Invest Clin 65(3): 267 - 278, 2024 https://doi.org/10.54817/IC.v65n3a01
Corresponding authors: Shaoqun Liu and Chang Su. Department of Gastrointestinal Surgery, Minhang Hospital
Affiliated to Fudan University, Xinzhuang Town, Shanghai 201199, China. E-mail: liushaoqun@fudan.edu.cn
E-mail: surgeon_suchang@fudan
Dihydroartemisinin, an artemisinin
derivative, reverses oxaliplatin resistance
in human colorectal cancer cells
by regulating the SIRT3/PI3K/AKT
signalling pathway.
Xiaodong Shen, Chencheng Shi, Ming Lei, Rongjian Zhou, Shaoqun Liu and Chang Su
Department of Gastrointestinal Surgery, Minhang Hospital Affiliated to Fudan
University, Shanghai, China.
Keywords: dihydroartemisinin; SIRT3; oxaliplatin; colorectal cancer.
Abstract. Dihydroartemisinin (DHA), a derivative of artemisinin, has been
shown to act as a chemosensitizer of various cancer chemotherapeutic agents
both in vitro and in vivo. However, in colorectal cancer (CRC), no study has fo-
cused on the effect of DHA on oxaliplatin (L-OHP) resistance. Our study aimed
to examine the effectiveness of DHA in reversing the resistance of human CRC
cells to L-OHP, as well as its underlying molecular mechanisms. LoVo cells were
purchased from ATCC, while LoVo/L-OHP cells were obtained by exposing LoVo
cells to progressively increasing concentrations of L-OHP. LoVo/L-OHP were
treated with various concentrations of DHA, and cell apoptosis ratio and vi-
ability were assessed by flow cytometry and CCK-8. Our results showed that
DHA treatment remarkably decreased the viability of LoVo/L-OHP cells and
increased the apoptosis ratio. As the mechanism of action, we found that DHA
enhanced the expression of Sirtuin 3 (SIRT3) and suppressed the phosphati-
dylinositol 3-kinase (PI3K)/AKT signalling cascade. Silencing of SIRT3 reversed
the effect of DHA on cell apoptosis and viability by activating the PI3K/AKT
axis in LoVo/L-OHP cells. Overall, our study found that DHA has the ability to
counteract L-OHP resistance in LoVo/L-OHP cells through the modulation of
the SIRT3/PI3K/AKT signalling pathway, suggesting a new research target for
CRC treatment.
268 Shen et al.
Investigación Clínica 65(3): 2024
La dihidroartemisinina, un derivado de la artemisinina, revierte
la resistencia al oxaliplatino en células humanas de cáncer
colorrectal mediante la regulación de la vía de señalización
SIRT3/PI3K/AKT.
Invest Clin 2024; 65 (3): 267 – 278
Palabras clave: dihidroartemisinina; SIRT3; oxaliplatino; cáncer colorrectal.
Resumen. Se ha demostrado que la dihidroartemisinina (DHA), un deri-
vado de la artemisinina, actúa como quimiosensibilizador de diversos agentes
quimioterapéuticos contra el cáncer tanto in vitro como in vivo. Sin embargo,
en el cáncer colorrectal (CCR), ningún estudio se ha centrado en el efecto del
DHA sobre la resistencia al oxaliplatino (L-OHP). El objetivo de nuestro estu-
dio era examinar la eficacia del DHA para revertir la resistencia de las células
humanas de CCR al L-OHP, así como sus mecanismos moleculares subyacentes.
Las células LoVo se adquirieron a ATCC, mientras que las células LoVo/L-OHP
se obtuvieron exponiendo las células LoVo a concentraciones progresivamente
crecientes de L-OHP. Las células LoVo/L-OHP se trataron con diversas con-
centraciones de DHA, y el índice de apoptosis celular y la viabilidad se evalua-
ron mediante citometría de flujo y CCK-8. Nuestros resultados mostraron que
el tratamiento con DHA disminuía notablemente la viabilidad de las células
LoVo/L-OHP y aumentaba el índice de apoptosis. Desde el punto de vista de
su mecanismo de acción, se observó que el DHA potenciaba la expresión de la
sirtuina 3 (SIRT3) y suprimía la cascada de señalización fosfatidilinositol 3-ci-
nasa (PI3K)/AKT. El silenciamiento de SIRT3 revirtió el efecto del DHA sobre la
apoptosis y la viabilidad celular a través de la activación del eje PI3K/AKT en las
células LoVo/L-OHP. En conjunto, nuestro estudio descubrió la capacidad del
DHA para contrarrestar la resistencia a L-OHP en células LoVo/L-OHP a través
de la modulación de la vía de señalización SIRT3/PI3K/AKT, lo cual sugiere una
nueva foco de investigación para el tratamiento del CCR.
Received: 25-07-2023 Accepted: 27-09-2023
INTRODUCTION
According to the 2020 GLOBOCAN sta-
tistics on the incidence and mortality rates
of 36 types of cancer worldwide, colorectal
cancer (CRC) ranks third in terms of inci-
dence (10.0%) but second in terms of mor-
tality rate (9.4%) 1. Chemotherapy based on
oxaliplatin (L-OHP) is a crucial component
of the comprehensive treatment for CRC,
which can prolong the survival of patients
and improve their quality of life 2-4. How-
ever, primary and/or acquired resistance to
L-OHP remains a significant factor limiting
the effectiveness of CRC treatment 5,6. There-
fore, it is urgent to clarify the mechanisms
of L-OHP resistance in CRC and to find cor-
responding resistance reversal agents or
chemosensitizers.
Previous studies have demonstrated
that the mechanisms of resistance to plati-
num-based chemotherapy drugs may be re-
Dihydroartemisinin reverses oxaliplatin resistance in colorectal cancer 269
Vol. 65(3): 267 - 278, 2024
lated to the abnormal regulation of signal-
ling networks within tumor cells caused by
the sustained action of drugs. For example,
abnormalities in signalling pathways such as
PI3K/AKT and NF-κB give the cells strong
anti-apoptotic abilities, leading to resistance
to chemotherapy drugs 7,8. Sirtuin 3 (Sirt3)
plays a crucial role in controlling physiologi-
cal and pathological processes, such as regu-
lating inflammation, oxidative stress, cell
proliferation, differentiation, and apoptosis.
This protein belongs to the sirtuin protein
family, known for its diverse biological func-
tions 9-11. Research has found that SIRT3 can
cause mutant p53 to be deacetylated, lead-
ing to cancer cell apoptosis and increased
sensitivity to chemotherapeutic drugs 12.
Other evidence also suggested that SIRT3
could improve disease conditions by regu-
lating apoptosis and utilizing the PI3K/Akt
pathway in various diseases 13,14. Dihydroarte-
misinin (DHA) is a derivative of artemisinin,
and there is evidence that DHA can act as
a chemosensitizer for various cancer chemo-
therapeutic drugs both in vitro and in vivo15.
The latest research has found that DHA can
alleviate acute and chronic pain by increas-
ing the activity of SIRT3 16. However, in CRC,
no study has focused on the effect of DHA on
L-OHP resistance through the SIRT3/PI3K/
AKT axis.
Based on previous studies, we hypoth-
esize that DHA may affect the resistance of
human CRC cells to L-OHP by regulating the
SIRT3/PI3K/AKT axis. Our study aimed to
examine the effectiveness of DHA in revers-
ing the resistance of human colorectal can-
cer cells to L-OHP, as well as its underlying
molecular mechanisms.
METHOD
Cell culture and induction of L-OHP
resistance
Human CRC cells (LoVo, not resistant
to L-OHP) were obtained from the American
Type Culture Collection (CCL-229, ATCC)
in Manassas, Virginia. The human L-OHP-
resistant CRC cells (LoVo/L-OHP, resistant
to L-OHP) were obtained by exposing LoVo
cells to progressively increased concentra-
tions of L-OHP 17. The initial concentration
of L-OHP was 5 μM, and cells were exposed
to L-OHP for two days in each cycle, followed
by recovery in L-OHP-free culture medium.
The same concentration of L-OHP was re-
peated for three cycles. After completing
three cycles at the same concentration of
L-OHP stimulation, the dose was escalated.
This procedure was repeated until the final
concentration of L-OHP reached 70 μM.
Only cells that remained resistant to L-OHP
after being cultured in an L-OHP-free me-
dium for at least six months were used for
subsequent experiments.
The LoVo/L-OHP and LoVo cells were
cultured at 37°C and 5% CO2 in RPMI-1640
medium (Thermo Fisher Scientific, Inc.,
MA, USA) that was supplemented with 10%
Gibco® fetal bovine serum (FBS) and 100
μg/mL penicillin-streptomycin. Cells were
treated with PI3K inhibitor LY294002 (1μM)
for 24 hours to inhibit PI3K signaling.
Preparation of Dihydroartemisinin
Solution
A stock solution of dihydroartemisinin
(obtained from Puyi Biological, Nanjing,
China) was prepared by dissolving it in a
small quantity of dimethyl sulfoxide (DMSO,
procured from Sigma-Aldrich, USA). The
resulting stock solution was formulated to
have a concentration of either 100 or 300
nmol/L, which was sonicated and stored
at 4°C. In subsequent experiments, all cell
samples were treated with 30 μM L-OHP or
an equivalent amount phosphate-buffered
saline (PBS) for treatment.
Cell transfection
The siRNA targeting the SIRT3 gene (si-
SIRT3#1, #2, #3) and the negative control
(NC) were procured from GeneChem Corp.
for cell transfection experiments.
270 Shen et al.
Investigación Clínica 65(3): 2024
According to the manufacturer’s in-
structions, the LoVo/L-OHP cells were trans-
fected with 5 nM of the aforementioned si-
RNA or negative controls. Transfection was
carried out by using the lipofectamine 3000
(Invitrogen) reagent, following the pre-
scribed protocol, while incubating the cells
in a serum-free Opti-MEM medium. After 48
hours of transfection, the transfection effi-
ciency was evaluated through RT-qPCR.
Cell Counting Kit-8 (CCK-8)
LoVo, LoVo/L-OHP, and LoVo/L-
OHP+DHA (100nmol/L or 300nmol/L)
cells were treated with different concentra-
tions of OHP (5μM-70μM) 17,18. Cells were
cultured into 96-well plates at 5×103 cells/
well density. After incubating the cells with
CCK-8 reagent (10μL, Sangon) for 2 hours,
the absorbance at 450 nm was determined
on a microplate reader (Thermo Fisher Sci-
entific, MA, USA). The half-maximal inhibi-
tory concentration (IC50) of L-OHP was de-
termined in LoVo and LoVo/L-OHP cells 19.
Reverse transcription-quantitative
polymerase chain reaction (RT-qPCR)
RT-qPCR was used to determine the ex-
pression of PI3K, SIRT3 and AKT in the cells.
We utilized the RNAiso plus kit (procured
from Takara, Japan) to obtain RNA from
cells and subsequently performed reverse
transcription into cDNA using the Prime-
ScriptTM one-step qRT-PCR kit (obtained
from Takara, Dalian, China). For RT-qPCR,
we employed the SYBR Premix ExTaq (TaKa-
Ra) and the ABI PRISM7300 Sequence De-
tection System (from Applied Biosystems).
Primer sequences are shown in Table 1. GAP-
DH served as an internal control. The mRNA
expressions were calculated by the 2-ΔΔCt
method.
Western blot
Total proteins were extracted from the
cell lysates and quantified using the Pierce™
BCA Protein Assay Kit (Thermo Scientific).
Then, 20 μg of the extracted proteins were
separated by SDS-PAGE and transferred onto
PVDF membranes. The PVDF membranes
were then incubated with a primary antibody
at 4˚C overnight. After washing, the mem-
branes were incubated with Goat Anti-Rab-
bit IgG H&L secondary antibody (ab96899,
1/1000) at 37˚C for 45 minutes. The prima-
ry antibodies included the SIRT3 antibody
ab217319, 1/1000, Abcam), PI3K antibody
(ab140307, 1/2000, Abcam), AKT antibody
(ab8805, 1/1000, Abcam), bcl-2 antibody
(ab32124, 1/1000, Abcam), Bax antibody
(ab32503, 1/1000, Abcam), cleaved cas-
pase-3 antibody (ab32042, 1/5000, Abcam),
P-gb (ab202976, 1/1000, Abcam), GAPDH
antibody (ab8245, 1/1000, Mybiosource),
and GAPDH was served as an internal con-
trol. We utilized the Pierce ECL Western
Blotting Substrate to visualize the protein
bands, which was procured from Pierce in
Shanghai, China. The membranes were de-
veloped and imaged using a chemilumines-
cence imaging system.
Flow cytometry for cell apoptosis
Cells were incubated at 37°C with 5%
CO2. After the incubation of 24 h, LoVo/L-
OHP cells were collected into 1.5 mL tubes
with annexin-FITC and propidium iodide
(PI) regents, followed by incubation for 10
min. To detect cell apoptosis, 200 μL PI was
supplemented with 1 mL phosphate buffer
Table 1
The sequences of all constructs.
Name Sequence
SIRT3 Forward 5’-CCCCAAGCCCTTTTTCACTTT-3’
Reverse 5’-CGACACTCTCTCAAGCCCA-3’
PI3K Forward 5’-TCTAAACCCTGCTCATC-3’
Reverse 5’-CTTGCCGTAAATCATCCC-3’
AKT Forward 5’-CTACAACCAGGACCATGAGAAG-3’
Reverse 5’-ACACGATACCGGCAAAGAAG-3’
GAPDH Forward 5-CACCCACTCCTCCACCTTTG-3
Reverse 5-CCACCACCCTGTTGCTGTAG-3
Dihydroartemisinin reverses oxaliplatin resistance in colorectal cancer 271
Vol. 65(3): 267 - 278, 2024
solution (PBS) into the flow tube. Then, the
apoptotic cells were measured using flow cy-
tometry (Beckman Coulter).
Statistical analysis
The data is presented as mean ± SD,
and we utilized one-way ANOVA followed by
a Tukey post hoc test for statistical compari-
sons. For significance, a p-value less than
0.05 was considered.
RESULTS
DHA inhibited the proliferation of LoVo/
L-OHP cells and induced their apoptosis
We first treated LoVo/L-OHP cells with
different concentrations (100 nmol/L or 300
nmol/L) of DHA then used the CCK-8 assay to
detect the IC50 of all cells. As shown in Fig.
1-A, the IC50 of LoVo/L-OHP cells to L-OHP
remarkably declined (p<0.05) after DHA
treatment. In the next experiment, we treat-
ed LoVo/L-OHP cells with 30μM L-OHP or an
equal amount of phosphate-buffered saline
(PBS) and investigated the changes in cell vi-
ability of LoVo/L-OHP cells after DHA treat-
ment. Our findings demonstrated a notable
decline in cell viability of LoVo/L-OHP cells
following DHA treatment (p<0.05, as de-
picted in Fig. 1-B). Moreover, flow cytometry
analysis substantiated a marked rise in apop-
totic cells in LoVo/L-OHP cells when treated
with 300 nmol/L DHA (p<0.05, as illustrated
in Fig. 1-C). Furthermore, compared with the
LoVo/L-OHP group, the expression of cleaved
caspase-3 protein and the Bax/Bcl-2 ratio in
the L-OHP +300 nmol/L DHA and L-OHP
+100nmol/L DHA groups were remarkably
increased, while the expression of P-gb pro-
tein was markedly decreased (p<0.05, Fig.
1-D). In the subsequent experiments, cells
were treated with 300 nmol/L of DHA.
The expression of SIRT3, PI3K, and AKT
in LoVo/L-OHP cells after DHA treatment
To further investigate the expression of
SIRT3, PI3K, and AKT in LoVo/L-OHP cells
after DHA treatment, we examined the ex-
pression of SIRT3, PI3K, and AKT in each
group of cells using WB and PCR. As shown in
Figs 2 A-B, our results indicated a significant
decrease in SIRT3 expression level, whereas
PI3K and AKT expression levels were signifi-
cantly elevated in LoVo/L-OHP cells com-
pared to the LoVo group (p<0.05). After ad-
ministering L-OHP to LoVo/L-OHP cells and
subsequently treating them with 300 nmol/L
DHA, we observed a noteworthy upsurge
in the protein and mRNA levels of SIRT3 as
compared to the LoVo/L-OHP+L-OHP group
(p<0.05). Furthermore, we observed a reduc-
tion in expression levels of PI3K and AKT in
cells following DHA treatment (p<0.05).
Silencing SIRT3 promoted L-OHP
resistance in LoVo/L-OHP cells after DHA
treatment through the PI3K/AKT pathway
To validate the role of the SIRT3/PI3K/
AKT signalling pathway in L-OHP resistance
of LoVo/L-OHP cells after DHA treatment,
we transfected cells with si-SIRT3#1, si-
SIRT3#2, si-SIRT3#3, and si-NC and mea-
sured the knockdown efficiency of SIRT3.
As shown in Figs. 3 A-B, in LoVo/L-OHP
cells after DHA treatment, si-SIRT3#1,
si-SIRT3#2, and si-SIRT3#3 all markably
reduced the expression of SIRT3, and we
selected si-SIRT3#2 with the highest knock-
down efficiency for subsequent experiments.
In addition, we also used the PI3K inhibitor
LY294002 to inhibit the PI3K/AKT path-
way and divided the cells into groups: conc-
trl (LoVo/L-OHP+L-OHP+DHA), si-NC
(conctrol+si-NC), si-SIRT3 (conctrol+si-
SIRT3), and si-SIRT3+LY294002 (conctrol+
si-SIRT3+LY294002). As shown in Fig. 3-C,
compared with the si-NC group, silencing
SIRT3 remarkably increased cell viability
and reduced the proportion of cell apopto-
sis (p<0.05). However, the addition of the
PI3K agonist LY294002 reversed this effect.
Compared with the si-SIRT3 group, the si-
SIRT3+LY294002 group had significantly
reduced cell viability and increased apopto-
sis (Fig. 3-D).
272 Shen et al.
Investigación Clínica 65(3): 2024
Effects of SIRT3 knockdown on relevant
proteins in LoVo/L-OHP cells after DHA
treatment
Finally, we used WB to measure the
protein expression of the SIRT3/PI3K/AKT
signalling pathway proteins, the drug-resis-
tant protein P-gb, and the apoptosis-related
proteins cleaved caspase-3, Bax and Bcl-2. As
illustrated in Fig. 4, our data demonstrated
that transfection with si-SIRT3 resulted in a
notable decline in SIRT3 protein expression
and increased protein expression of PI3K
and AKT (p<0.05).
Fig. 1. DHA inhibited the proliferation of LoVo/L-OHP cells and induced their apoptosis. (A) The IC50 of
all groups of cells were detected by CCK-8 assay. (B) Cell viability of all groups of cells was detected
by CCK-8 assay. (C) The apoptosis rate of all groups of cells was analyzed by flow cytometry. (D) The
expression of P-gp protein related to drug resistance and apoptosis-related proteins, cleaved caspase-3
protein, Bax, and Bcl-2, was detected by Western blot. Continuous data were compared using ANOVA.
*p<0.05 compared with the L-OHP group. #p<0.05 compared with the LoVo/L-OHP group.
Dihydroartemisinin reverses oxaliplatin resistance in colorectal cancer 273
Vol. 65(3): 267 - 278, 2024
On the other hand, treatment with
PI3K inhibitor LY294002 remarkably re-
duced the protein expression levels of PI3K
and AKT (p<0.05). Silencing SIRT3 marked-
ly enhanced the expression of P-gb while re-
ducing the protein expression of cleaved cas-
pase-3 and the ratio of Bax/Bcl-2. However,
the PI3K inhibitor LY294002 reversed this
effect. In the si-SIRT3+LY294002 group, the
protein expression of P-gb decreased, while
the expression of cleaved caspase-3 and the
ratio of Bax/Bcl-2 significantly increased
(p<0.05).
DISCUSSION
For researchers, the mechanism of L-
OHP resistance in CRC is still elusive. De-
spite many molecular medicine studies to
explain the resistance mechanisms of CRC,
it is challenging to distinguish genuinely
effective targets for regulating resistance.
In addition, most existing multidrug resis-
tance reversal agents have issues such as
incomplete resistance reversal, high tech-
nical requirements, and significant adverse
reactions 20, 21. Therefore, finding new sensi-
tizers to combine with L-OHP to treat CRC
patients is still necessary. We demonstrated
that DHA could inhibit the L-OHP resistance
in LoVo human colorectal cancer cells by
regulating the SIRT3/PI3K/AKT axis.
Studies conducted in the past have dem-
onstrated that DHA can hinder the growth
of cancer cells and trigger apoptosis in pan-
creatic cancer, ovarian cancer, and colorec-
tal cancer. An added advantage of DHA is its
low toxicity towards normal tissue cells 22,
23. In addition, DHA also has a killing effect
on multidrug-resistant tumor cells 24. In our
study, we treated CRC-resistant cells LoVo/
L-OHP with different concentrations of DHA
and found that the viability of LoVo/L-OHP
cells marked decrease, and the proportion
of apoptotic cells increased significantly af-
ter DHA treatment. This phenomenon was
Fig. 2. The expression of SIRT3, PI3K and AKT in LoVo/L-OHP cells after DHA treatment. The SIRT3,
PI3K and AKT expression in each group were measured by WB (A) and PCR (B). ANOVA was used to
compare the groups. #p<0.05 compared with the LoVo/L-OHP group. *p<0.05 compared with the
LoVo/L-OHP+L-OHP group.
274 Shen et al.
Investigación Clínica 65(3): 2024
more pronounced in the group with higher
concentrations of DHA, indicating that DHA
can inhibit the resistance of LoVo/L-OHP to
L-OHP. Similar results have been obtained in
other cancers. In non-small-cell lung cancer
(NSCLC), DHA can eliminate radiation re-
sistance of NSCLC cell lines and xenograft
models by inhibiting the TGF-β, PI3K/Akt,
and STAT3 signalling pathways, and enhance
the anti-tumor effect of radiotherapy 25. In
breast cancer, DHA regulates the STAT3/
DDA1 axis to inhibit the proliferation of
breast cancer tumor cells 26. In addition, Yao
et al. confirmed that DHA can effectively sen-
sitize CRC cells to 5-fluorouracil (5-FU) by
mediating ROS-induced apoptosis 27. Wang
et al. also showed that DHA inhibits the char-
acteristics of cancer stem-like cells (CSLCs)
Fig. 3. Silencing SIRT3 promotes L-OHP resistance in LoVo/L-OHP cells after DHA treatment through
the PI3K/AKT pathway. The expression of SIRT3 was measured by WB (A) and PCR (B). (C) Cell via-
bility of all groups of cells was detected by CCK-8 assay. (D) The apoptosis rate of all groups of cells
was analyzed by flow cytometry. ANOVA was used to compare the groups. *p<0.05 vs si-NC group.
#p<0.05 vs si-SIRT3 group.
Dihydroartemisinin reverses oxaliplatin resistance in colorectal cancer 275
Vol. 65(3): 267 - 278, 2024
in CRC through the AKT/mTOR signalling
pathway 28. These results suggested that
DHA can be a sensitizing cancer treatment
agent.
The protein and mRNA levels of SIRT3
were enhanced in LoVo/L-OHP cells after
treatment with DHA, indicating that SIRT3
may be involved in the sensitization of LoVo/
L-OHP by DHA. Cao et al. found that acti-
vation of SIRT3 inhibits cisplatin resistance
in lung cancer cells and reduces the prolif-
eration and invasion of lung cancer cells 29.
Another research showed that the activity of
SIRT3 in colon cancer cells can be enhanced
by nitric oxide synthase (NOS), which helps
to promote cell apoptosis 30. To study the
mechanism of action, we silenced SIRT3 and
found that the viability of LoVo/L-OHP cells
improved, and the proportion of apoptotic
cells and apoptotic proteins cleaved cas-
pase-3 and the ratio of bax/bcl-2 decreased
significantly, indicating that the knockdown
of SIRT3 affected the resistance of LoVo/L-
OHP cells to L-OHP after DHA treatment.
Furthermore, we confirmed that silencing
SIRT3 enhanced the expression of PI3K and
AKT in LoVo/L-OHP cells. The aberrant acti-
vation of the PI3K/AKT pathway acts against
chemotherapy-induced apoptosis by increas-
ing the expression of anti-apoptotic genes
(bcl-2 and XIAP) and reducing the expres-
sion of pro-apoptotic genes (bax) 31. In our
study, we inhibited the PI3K/AKT axis with
LY294002 and found that the viability of
LoVo/L-OHP cells decreased while apopto-
sis increased. This also reversed the effect
of knocking down SIRT3 on the viability and
apoptosis of LoVo/L-OHP cells. Zhang et al.
32 found that inhibiting the PI3K/AKT signal-
ling pathway in CRC cells can inhibit cell
proliferation and activate apoptosis, thereby
reversing oxaliplatin resistance, consistent
with our research results.
Limitation
Our research also has some limitations.
Whether other genes regulate SIRT3 is not
clear. We also did not perform in vivo experi-
ments to confirm the apoptosis results. All
these need further studies to illustrate.
Our study found that DHA could inhibit
the L-OHP resistance in LoVo human colorec-
tal cancer cells by regulating the SIRT3/
PI3K/AKT axis. The results might provide a
new integrated treatment approach for CRC
treatment in clinical practice.
Ethics approval and consent to participate
The ethic approval was obtained from
the Ethic Committee of The Minhang Hos-
pital, Affiliated with Fudan University, and
Fig. 4. Effects of SIRT3 knockdown on relevant proteins in LoVo/L-OHP cells after DHA treatment. The
protein expression of SIRT3, PI3K, AKT, P-gb, cleaved caspase-3, Bax and Bcl-2 were measured by WB.
ANOVA was used to compare the groups. *p<0.05 vs si-NC group. #p<0.05 vs si-SIRT3 group.
276 Shen et al.
Investigación Clínica 65(3): 2024
written informed consent was obtained from
all patients.All of the authors have Consent-
ed to publish this research.
Availability of data and materials
The data are free to access and avail-
able upon request.
Competing interests
All authors declare no conflict of interest.
Funding
Project of Health Commission of
Minhang District, Shanghai, Grant No.
2021MW23.
ORCID number of authors
Xiaodong Shen:
0009-0000-0520-8896
Chencheng Shi:
0009-0008-1369-2946
Ming Lei:
0009-0000-8480-197X
Rongjian Zhou:
0009-0007-5598-0834
Shaoqun Liu:
0000-0002-2016-0947
Chang Su:
0009-0004-6481-2638
Authors’ contributions
Each author has made an important sci-
entific contribution to the study and assist-
ed with drafting or revising the manuscript.
REFERENCES
1. Sung H, Ferlay J, Siegel RL, Laversanne
M, Soerjomataram I, Jemal A, Bray F.
Global cancer statistics 2020: GLOBO-
CAN estimates of incidence and mortality
worldwide for 36 cancers in 185 countries.
CA Cancer J Clin 2021;71(3):209-249.
2. Cvitkovic E, Bekradda M. Oxaliplatin: a
new therapeutic option in colorectal can-
cer. Semin Oncol 1999;26(6):647-662.
3. Rovers KP, Wassenaar ECE, Lurvink
RJ, Creemers G-JM, Burger JWA, Los
M, Huysentruyt CJR, van Lijnschoten
G, Nederend J, Lahaye MJ. Pressurized
intraperitoneal aerosol chemotherapy
(oxaliplatin) for unresectable colorectal
peritoneal metastases: a multicenter, sin-
gle-arm, phase II trial (CRC-PIPAC). Ann
Surg Oncol 2021: 28(9):5611-5616.
4. Ranieri G, Laforgia M, Nardulli P, Fe-
rraiuolo S, Molinari P, Marech I, Gada-
leta CD. Oxaliplatin-based intra-arterial
chemotherapy in colorectal cancer li-
ver metastases: a review from pharma-
cology to clinical application. Cancers
2019;11(2):141.
5. Zhang Y, Li C, Liu X, Wang Y, Zhao R,
Yang Y, Zheng X, Zhang Y, Zhang X. cir-
cHIPK3 promotes oxaliplatin-resistance
in colorectal cancer through autopha-
gy by sponging miR-637. EBioMedicine
2019;48:277-288.
6. Lai M, Liu G, Li R, Bai H, Zhao J, Xiao
P, Mei J. Hsa_circ_0079662 induces the
resistance mechanism of the chemothe-
rapy drug oxaliplatin through the TNF-α
pathway in human colon cancer. J Cell Mol
Med 2020;24(9):5021-5027.
7. Mitsuuchi Y, Johnson SW, Selvakumaran
M, Williams SJ, Hamilton TC, Testa JR.
The phosphatidylinositol 3-kinase/AKT
signal transduction pathway plays a critical
role in the expression of p21WAF1/CIP1/
SDI1 induced by cisplatin and paclitaxel.
Cancer Research 2000;60(19):5390-5394.
8. Selfe J, Goddard NC, McIntyre A, Ta-
ylor KR, Renshaw J, Popov SD, Thway
K, Summersgill B, Huddart RA, Gilbert
DC. IGF1R signalling in testicular germ
cell tumour cells impacts on cell survival
and acquired cisplatin resistance. J Pathol
2018;244(2):242-253.
9. Lee S, Jeon Y-M, Jo M, Kim H-J. Overex-
pression of SIRT3 suppresses oxidative
stress-induced neurotoxicity and mito-
chondrial dysfunction in dopaminergic neu-
ronal cells. Exp Neurobiol 2021;30(5):341.
Dihydroartemisinin reverses oxaliplatin resistance in colorectal cancer 277
Vol. 65(3): 267 - 278, 2024
10. Dikalova AE, Pandey A, Xiao L, Arslanbae-
va L, Sidorova T, Lopez MG, Billings Iv
FT, Verdin E, Auwerx J, Harrison DG. Mi-
tochondrial deacetylase Sirt3 reduces vas-
cular dysfunction and hypertension while
Sirt3 depletion in essential hypertension is
linked to vascular inflammation and oxida-
tive stress. Circ Res 2020;126(4):439-452.
11. Gao J, Feng Z, Wang X, Zeng M, Liu J,
Han S, Xu J, Chen L, Cao K, Long J.
SIRT3/SOD2 maintains osteoblast diffe-
rentiation and bone formation by regula-
ting mitochondrial stress. Cell Death Di-
ffer 2018;25(2):229-240.
12. Guo R, Li Y, Xue Y, Chen Y, Li J, Deng X, Su
J, Liu Y, Sun L. SIRT3 increases cisplatin
sensitivity of small-cell lung cancer through
apoptosis. Gene 2020;745:144629.
13. Xu K, He Y, Moqbel SAA, Zhou X, Wu
L, Bao J. SIRT3 ameliorates osteoarthri-
tis via regulating chondrocyte autopha-
gy and apoptosis through the PI3K/Akt/
mTOR pathway. Int J Biol Macromol
2021;175:351-360.
14. Wang Z, Li Y, Wang Y, Zhao K, Chi Y,
Wang B. Pyrroloquinoline quinine protects
HK-2 cells against high glucose-induced
oxidative stress and apoptosis through
Sirt3 and PI3K/Akt/FoxO3a signalling
pathway. Biochem Biophys Res Commun
2019;508(2):398-404.
15. Li Q, Ma Q, Cheng J, Zhou X, Pu W,
Zhong X, Guo X. Dihydroartemisinin as a
sensitizing agent in cancer therapies. On-
coTargets Ther 2021;14:2563.
16. Zhang L, Wang Z, Wang C, Li J, Zhao Y,
Zhang H, Yu Y, Wang G, Li Y. Dihydroarte-
misinin protects against acute and chronic
pain via SIRT3-dependent inhibition of pe-
roxiredoxin-3 hyperacetylation and pero-
xynitrite accumulation in mice. Research
Square 2022. https://doi.org/10.21203/
rs.3.rs-1607556/v1
17. Wang Q, Wei J, Wang C, Zhang T, Huang
D, Wei F, He F, Cai W, Yang P, Zeng S.
Gambogic acid reverses oxaliplatin resis-
tance in colorectal cancer by increasing
intracellular platinum levels. Oncol Lett
2018;16(2):2366-2372.
18. Wang C-H, Baskaran R, Ng SS-C, Wang
T-F, Li C-C, Ho T-J, Hsieh DJ-Y, Kuo C-H,
Chen M-C, Huang C-Y. Platycodin D con-
fers oxaliplatin resistance in colorectal
cancer by activating the LATS2/YAP1 axis
of the hippo signalling pathway. J Cancer
2023;14(3):393.
19. Liu X, Wu B, Chen H, Sun H, Guo X, Sun
T, Zhou D, Yang S. Intense endoplasmic
reticulum stress (ERS)/IRE1α enhanced
Oxaliplatin efficacy by decreased ABCC10
in colorectal cancer cells. BMC Cancer
2022;22(1):1369.
20. Krishna R, Mayer LD. Multidrug resistan-
ce (MDR) in cancer: mechanisms, reversal
using modulators of MDR and the role of
MDR modulators in influencing the phar-
macokinetics of anticancer drugs. Eur J
Pharm Sci 2000;11(4):265-283.
21. Liang Y, Zhu D, Zhu L, Hou Y, Hou L,
Huang X, Li L, Wang Y, Li L, Zou H. Di-
chloroacetate overcomes oxaliplatin che-
moresistance in colorectal cancer through
the miR-543/PTEN/Akt/mTOR pathway. J
Cancer 2019;10(24):6037.
22. Aung W, Sogawa C, Furukawa T, Saga T.
Anticancer effect of dihydroartemisinin
(DHA) in a pancreatic tumor model evalua-
ted by conventional methods and optical
imaging. Anticancer Res 2011;31(5):1549-
1558.
23. Jiao Y, Ge Cm, Meng Qh, Cao Jp, Tong J,
Fan Sj. Dihydroartemisinin is an inhibitor
of ovarian cancer cell growth 1. Acta Phar-
macol Sin 2007;28(7):1045-1056.
24. Michaelis M, Kleinschmidt MC, Barth S,
Rothweiler F, Geiler J, Breitling R, Ma-
yer B, Deubzer H, Witt O, Kreuter J. Anti-
cancer effects of artesunate in a panel of
chemoresistant neuroblastoma cell lines.
Biochem Pharmacol 2010;79(2):130-136.
25. Zhang H, Zhou F, Wang Y, Xie H, Luo S,
Meng L, Su B, Ye Y, Wu K, Xu Y. Elimi-
nating radiation resistance of non-small
cell lung cancer by dihydroartemisinin
through abrogating immunity escaping
and promoting radiation sensitivity by in-
hibiting PD-L1 expression. Front Oncol
2020;10:595466.
278 Shen et al.
Investigación Clínica 65(3): 2024
26. Zhang J, Li Y, Wang J-G, Feng J-Y, Huang
G-D, Luo C-G. Dihydroartemisinin affects
STAT3/DDA1 signalling pathway and re-
verses breast cancer resistance to cispla-
tin. Am J Chin Med 2023;51(02):445-459.
27. Yao Z, Bhandari A, Wang Y, Pan Y, Yang
F, Chen R, Xia E, Wang O. Dihydroarte-
misinin potentiates antitumor activity of
5-fluorouracil against a resistant colorec-
tal cancer cell line. Biochem Biophys Res
Commun 2018;501(3):636-642.
28. Wang Y, Yang Z, Zhu W, Chen Y, He X, Li J,
Han Z, Yang Y, Liu W, Zhang K. Dihydroar-
temisinin inhibited stem celllike proper-
ties and enhanced oxaliplatin sensitivity
of colorectal cancer via AKT/mTOR signa-
lling. Drug Dev Res 2023;84(5):988-998.
29. Cao Y, Li P, Wang H, Li L, Li Q. SIRT3 promo-
tion reduces resistance to cisplatin in lung
cancer by modulating the FOXO3/CDT1
axis. Cancer Med 2021;10(4):1394-1404.
30. Wang Q, Ye S, Chen X, Xu P, Li K, Zeng
S, Huang M, Gao W, Chen J, Zhang Q.
Mitochondrial NOS1 suppresses apoptosis
in colon cancer cells through increasing
SIRT3 activity. Biochem Biophys Res Com-
mun 2019;515(4):517-523.
31. Liu R, Chen Y, Liu G, Li C, Song Y, Cao
Z, Li W, Hu J, Lu C, Liu Y. PI3K/AKT
pathway as a key link modulates the multi-
drug resistance of cancers. Cell Death Dis
2020;11(9):797.
32. Zhang Y, Xu Z, Sun Y, Chi P, Lu X. Knoc-
kdown of KLK11 reverses oxaliplatin resis-
tance by inhibiting proliferation and acti-
vating apoptosis via suppressing the PI3K/
AKT signal pathway in colorectal cancer
cell. OncoTargets Ther 2018:809-821.