ArticlePDF Available

Targeting the ERK1/2 and p38 MAPK pathways attenuates Golgi tethering factor golgin-97 depletion-induced cancer progression in breast cancer

Authors:

Abstract and Figures

Background The Golgi apparatus is widely considered a secretory center and a hub for different signaling pathways. Abnormalities in Golgi dynamics can perturb the tumor microenvironment and influence cell migration. Therefore, unraveling the regulatory network of the Golgi and searching for pharmacological targets would facilitate the development of novel anticancer therapies. Previously, we reported an unconventional role for the Golgi tethering factor golgin-97 in inhibiting breast cell motility, and its downregulation was associated with poor patient prognosis. However, the specific role and regulatory mechanism of golgin-97 in cancer progression in vivo remain unclear. Methods We integrated genetic knockout (KO) of golgin-97, animal models (zebrafish and xenograft mice), multi-omics analysis (next-generation sequencing and proteomics), bioinformatics analysis, and kinase inhibitor treatment to evaluate the effects of golgin-97 KO in triple-negative breast cancer cells. Gene knockdown and kinase inhibitor treatment followed by qRT‒PCR, Western blotting, cell viability, migration, and cytotoxicity assays were performed to elucidate the mechanisms of golgin-97 KO-mediated cancer invasion. A xenograft mouse model was used to investigate cancer progression and drug therapy. Results We demonstrated that golgin-97 KO promoted breast cell metastasis in zebrafish and xenograft mouse models. Multi-omics analysis revealed that the Wnt signaling pathway, MAPK kinase cascades, and inflammatory cytokines are involved in golgin-97 KO-induced breast cancer progression. Targeting the ERK1/2 and p38 MAPK pathways effectively attenuated golgin-97-induced cancer cell migration, reduced the expression of inflammatory mediators, and enhanced the chemotherapeutic effect of paclitaxel in vitro and in vivo. Specifically, compared with the paclitaxel regimen, the combination of ERK1/2 and p38 MAPK inhibitors significantly prevented lung metastasis and lung injury. We further demonstrated that hypoxia is a physiological condition that reduces golgin-97 expression in cancer, revealing a novel and potential feedback loop between ERK/MAPK signaling and golgin-97. Conclusion Our results collectively support a novel regulatory role of golgin-97 in ERK/MAPK signaling and the tumor microenvironment, possibly providing new insights for anti-breast cancer drug development. Supplementary Information The online version contains supplementary material available at 10.1186/s12964-024-02010-0.
Golgin-97 KO modulates the malignancy of breast cancer cells in vivo. A Representative images showing that MDA-MB-231 and golgin-97 KO cells were injected into the perivitelline cavity of zebrafish embryos and analyzed with a dissecting microscope. The location of the cancer cells is indicated in red on bright field images of zebrafish. The white arrows indicate dispersed breast cancer cells. B Quantification of the number of cell clusters on the basis of the acquired images in Fig. 1A. Each dot represents one live fish for image analysis. The quantitative data were analyzed via one-way ANOVA. C Representative images of lung metastases formed from MDA-MB-231 and 2-9 KO cell-derived NOD/SCID mice on day 28 after tail vein injection. D Representative IHC images with H&E staining of lung metastases. Scale bars, 100 μm. The yellow arrows delineate the tumor region. E Representative images showing the tumor sizes on day 42 after the injection of MDA-MB-231 and 2-9 KO cells into the right middle mammary fat pads of the mice. F Tumor growth was monitored at the indicated times after the injection of MDA-MB-231 and 2-9 KO cells, and tumor growth curves were constructed. The data were analyzed via two-way ANOVA. G Compared with those injected with MDA-MB-231 cells, NOD/SCID mice injected with golgin-97 KO cells exhibited poor survival (n = 9 mice per group). Kaplan-Meier plots of mouse survival using a standard tumor volume of 300 mm 3 as a criterion for euthanasia. P values were determined via the log-rank test. The data are presented as the means ± SDs (*p < 0.05; **p < 0.01; ****, P < 0.0001)
… 
Content may be subject to copyright.
Liuetal. Cell Communication and Signaling (2025) 23:22
https://doi.org/10.1186/s12964-024-02010-0
RESEARCH Open Access
© The Author(s) 2025. Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0
International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if
you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or
parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To
view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
Cell Communication
and Signaling
Targeting theERK1/2 andp38 MAPK
pathways attenuates Golgi tethering factor
golgin-97 depletion-induced cancer progression
inbreast cancer
Yu-Chin Liu1, Tsung-Jen Lin1,2, Kowit-Yu Chong3,4,5,6, Guan-Ying Chen1, Chia-Yu Kuo1, Yi-Yun Lin7,
Chia-Wei Chang7, Ting-Feng Hsiao7, Chih-Liang Wang8,9, Yo-Chen Shih7 and Chia-Jung Yu1,7,9,10*
Abstract
Background The Golgi apparatus is widely considered a secretory center and a hub for different signaling pathways.
Abnormalities in Golgi dynamics can perturb the tumor microenvironment and influence cell migration. Therefore,
unraveling the regulatory network of the Golgi and searching for pharmacological targets would facilitate the devel-
opment of novel anticancer therapies. Previously, we reported an unconventional role for the Golgi tethering factor
golgin-97 in inhibiting breast cell motility, and its downregulation was associated with poor patient prognosis. How-
ever, the specific role and regulatory mechanism of golgin-97 in cancer progression in vivo remain unclear.
Methods We integrated genetic knockout (KO) of golgin-97, animal models (zebrafish and xenograft mice), multi-
omics analysis (next-generation sequencing and proteomics), bioinformatics analysis, and kinase inhibitor treatment
to evaluate the effects of golgin-97 KO in triple-negative breast cancer cells. Gene knockdown and kinase inhibitor
treatment followed by qRTPCR, Western blotting, cell viability, migration, and cytotoxicity assays were performed
to elucidate the mechanisms of golgin-97 KO-mediated cancer invasion. A xenograft mouse model was used to inves-
tigate cancer progression and drug therapy.
Results We demonstrated that golgin-97 KO promoted breast cell metastasis in zebrafish and xenograft mouse mod-
els. Multi-omics analysis revealed that the Wnt signaling pathway, MAPK kinase cascades, and inflammatory cytokines
are involved in golgin-97 KO-induced breast cancer progression. Targeting the ERK1/2 and p38 MAPK pathways
effectively attenuated golgin-97-induced cancer cell migration, reduced the expression of inflammatory mediators,
and enhanced the chemotherapeutic effect of paclitaxel in vitro and in vivo. Specifically, compared with the paclitaxel
regimen, the combination of ERK1/2 and p38 MAPK inhibitors significantly prevented lung metastasis and lung injury.
We further demonstrated that hypoxia is a physiological condition that reduces golgin-97 expression in cancer, reveal-
ing a novel and potential feedback loop between ERK/MAPK signaling and golgin-97.
Conclusion Our results collectively support a novel regulatory role of golgin-97 in ERK/MAPK signaling
and the tumor microenvironment, possibly providing new insights for anti-breast cancer drug development.
Keywords Golgi, Golgin-97, Breast cancer, MAPK signaling, Metastasis, Inflammatory cytokine
*Correspondence:
Chia-Jung Yu
yucj1124@mail.cgu.edu.tw; yucj1124@gmail.com
Full list of author information is available at the end of the article
Page 2 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
Introduction
Breast cancer is the most common type of cancer in
female patients and the second leading cause of death
among women overall [1]. Triple-negative breast can-
cer (TNBC) has been shown to have a higher incidence
of metastasis, a poorer prognosis, and poorer overall
survival than other subtypes of breast cancer [2]. TNBC
lacks the expression of the sensitive therapeutic mark-
ers estrogen receptor, progesterone receptor, and human
epidermal growth factor receptor 2, leading to the inef-
fectiveness of endocrine therapy. Systemic chemotherapy
with anthracyclines and taxanes is the most common
first-line treatment modality [3]. However, anticancer
drug resistance, off-target toxicity, and complications,
including pneumonia with severe inflammation, are
major obstacles to the development of cancer chemo-
therapy regimens [46]. Although the possible molecu-
lar mechanisms of TNBC tumor growth, metastasis, and
therapy have been increasingly revealed, their clinical
application is still limited. us, identifying new molec-
ular targets for predicting clinical prognosis and finding
effective strategies for treating TNBC are crucial.
e Golgi apparatus is the trafficking hub that regu-
lates the anterograde transport of modified proteins and
lipids or retrieves extracellular proteins through retro-
grade transport from endosomes [7]. ere is growing
evidence that the Golgi apparatus is a hub for different
signaling pathways [810]. Several Golgi-localized pro-
teins have recently been reported to be involved in the
function of the Golgi and are related to the regulation of
cancer cell migration and invasion [10, 11]. Golgin-97,
localized at the trans-Golgi network (TGN), is a coiled-
coil protein first identified in Sjogren’s syndrome patients
[12]. Golgin-97 maintains Golgi integrity and acts as a
tethering molecule that mediates the vesicular traffick-
ing of specific cargoes (such as E-cadherin) [1315]. Gol-
gin-97 expression is significantly lower in invasive ductal
breast cancer than in ductal carcinoma insitu [16]. Low
expression of golgin-97 has also been positively associ-
ated with poor overall survival in patients with breast,
lung, and ovarian cancers, supporting a suppressive
role of golgin-97 in regulating cancer progression. We
then revealed a noncanonical role of golgin-97 in sup-
pressing TNBC cell motility through the modulation of
NF-κB activity in vitro [17]. Unfortunately, the inhibi-
tion of NF-κB alone is insufficient to induce pronounced
apoptosis in tumor cells, and long-term treatment with
NF-κB inhibitors influences normal cell function, lead-
ing to severe systemic toxicity and immunosuppression
[18]. Additionally, whether and how golgin-97 depletion
promotes cancer metastasis invivo and the mechanisms
affecting golgin-97 downregulation in breast cancer
remain unclear.
In this study, we established a cell line with stable
golgin-97 depletion via CRISPRCas9 gene knock-
out (KO) and demonstrated that golgin-97 depletion
promoted MDA-MB-231 cell migration and metasta-
sis. Multi-omics analysis and functional characteriza-
tion demonstrated the therapeutic potential of p38
MAPK inhibitors and ERK1/2 inhibitors in golgin-97
KO-induced cancer progression in vitro and in vivo,
confirming their biological importance in breast cancer
and anticancer drug development.
Materials andmethods
Cell culture, chemical reagents, andchemotherapy drugs
e TNBC cell line MDA-MB-231 was purchased from
the American Type Culture Collection (Manassas, Vir-
ginia). Golgin-97 KO MDA-MB-231 cells were gener-
ated via CRISPRCas9 gene KO in our laboratory. Both
wild-type MDA-MB-231 (hereafter referred to as MDA-
MB-231) cells and golgin-97 KO MDA-MB-231 cells
were cultured in Dulbecco’s modified Eagle’s medium
(DMEM) (Gibco, Waltham, MA, USA) containing 10%
FBS (Gibco), 1% penicillin/streptomycin (Gibco), and 1%
L-glutamine (Gibco). SB203580, U0126, and paclitaxel
were purchased from MedChemExpress (Monmouth
Junction, NJ, USA). Cobalt chloride (CoCl2) was pur-
chased from Sigma (St. Louis, MO, USA).
CRISPRCas9 gene deletion
Golgin-97/ KO MDA-MB-231 breast cancer cells were
generated with an optimized single-guide RNA (sgRNA;
human golgin-97, accession no. NM_002077.3) and the
Cas9 expression plasmid pRGEN-Cas9-CMV (ToolGen,
Seoul, Korea). Details for establishing cell lines with
golgin-97 KO are described in Appendix S1 (available
online).
Cancer metastasis inthezebrash model
e zebrafish xenotransplantation procedure was
described previously [19]. Briefly, 24 h postfertilization
(hpf), zebrafish embryos of the wild-type Tübingen strain
were dechorionated and anesthetized in E3 medium
containing phenylthiourea and tricaine before cell injec-
tion. MDA-MB-231 cells (control or golgin-97 KO) were
labeled with the lipophilic fluorochrome CM-DiI (Invit-
rogen, Waltham, MA, USA). Stained cells were injected
into the yolk sacs of zebrafish embryos via a microinjec-
tor according to the instruction manual. e injected
zebrafish were maintained in E3 medium. Images of the
zebrafish were acquired with a Z16 APO microscope
using a Leica DFC490 color camera system.
Page 3 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
Next‑generation sequencing (NGS) andbioinformatic
analysis
Total RNA was extracted from MDA-MB-231 and
golgin-97 KO cells via TRIzol Reagent (Invitrogen,
Waltham, MA, USA) according to the instruction man-
ual. NGS was carried out via Illumina’s sequencing-
by-synthesis technology (Illumina, USS) as described
previously [20]. For pathway enrichment analysis, the
differentially expressed genes (DEGs) whose expression
was altered twofold in golgin-97 KO cells compared
with that in MDA-MB-231 cells were subjected to path-
way analysis via DAVID Bioinformatics Resources 6.7
(https:// david-d. ncifc rf. gov/ home. jsp) and QIAGEN
Ingenuity Pathway Analysis (IPA) software.
Quantitative proteomic analysis ofthedierential
proteome resulting fromgolgin‑97 KO
Protein extracts prepared from MDA-MB-231 and gol-
gin-97 KO cells were digested with trypsin, chemically
labeled with a tandem mass tag (TMT), and then sub-
jected to two-dimensional liquid chromatographytan-
dem mass spectrometry (2D LCMS/MS). e details
for protein identification and quantification via mass
spectrometry are provided in Appendix S1 (available
online).
Small interfering RNA (siRNA) transfection
siRNAs targeting p38 MAPK, ERK1/2 and golgin-97
were purchased from ermo Fisher Scientific or
Dharmacon and used for gene knockdown with Lipo-
fectamine RNAiMAX reagents (Invitrogen, Grand
Island, NY, USA) according to the manufacturer’s
instructions. e sequences of the siRNAs used in this
study are listed in TableS1.
Cell viability assay
Cell viability was determined by using a Cell Counting
Kit-8 (CCK-8) assay (TEN-CCK8) (BIOTOOLS Co.,
Ltd., New Taipei City, Taiwan). e detailed methods
of the cell viability assay are available in Appendix S1
(available online).
Cell andanimal tissue RNA extraction andquantitative
RTPCR
Total RNA was extracted from cells or tumor tissue,
and cDNA was prepared for qRTPCR as described
previously [17]. e sequences of the primers used in
this study are listed in TableS2.
Western blotting
e detailed Western blotting methods are available
in Appendix S1. e primary antibodies used included
anti-golgin-97 (13192S), anti-IκBα (4814S), anti-p38
(8690S), anti-phospho-ERK1/2 (9101S), anti-cleaved
caspase-3 (#9661) and anti-caspase-3 (#9662), which
were purchased from Cell Signaling Technology (Dan-
vers, MA, USA). e anti-ERK (sc-93) antibody was
purchased from Santa Cruz (DT, USA). Anti-phospho-
p38 (09–272) and anti-β-actin (MAB1501) antibod-
ies were obtained from Merck Millipore (Burlington,
MA, USA). e anti-HIF1α (GTX127309) antibody was
purchased from GeneTex (Irvine, CA, USA). e anti-
TGN46 antibody was purchased from Bio-Rad (Hercu-
les, CA, USA).
Wound healing migration assay
e cell migration ability was analyzed via a wound heal-
ing assay. e detailed methods are available in Appendix
S1 (available online).
Animal model ofmetastasis
is study was approved by the Institutional Animal
Care and Use Committee of Chang Gung University
(CGU107–228). Four-week-old female immunodeficient
nonobese diabetic/SCID (NOD. CB17-Prkdcscid/JNarl)
mice were purchased from the National Laboratory
Animal Center (Taipei, Taiwan) to establish the tail vein
injection and orthotopic inoculation models as described
in Appendix S1 (available online).
Immunohistochemical (IHC) analysis
e detailed IHC methods are available in Appendix S1.
Statistical analysis
All experiments were performed at least in triplicate, and
the data are presented as the means ± standard devia-
tions (SDs). Prism 8.0 (GraphPad Software) statistical
software packages were used for data analysis. Unpaired t
tests, one-way ANOVA, two-way ANOVA, and log-rank
tests were used for comparisons between the control and
experimental groups. A P value less than 0.05 (P < 0.05)
was considered to indicate significance.
Results
Golgin‑97 KO modulates themalignancy ofbreast cancer
cells in vivo
Our previous studies revealed that golgin-97 knock-
down promotes NF-κB activation and leads to increased
migration/invasion of breast cancer cells [17]. Herein, we
applied CRISPR–Cas9 to generate four clonal golgin-97
KO cell lines (Fig. S1A). We confirmed that the expres-
sion levels of IκBα and TGN46 were reduced, accom-
panied by increased migration and invasion of 2–9 and
2–20 KO cells (Fig. S1B). erefore, to verify the effects
of golgin-97 depletion on cancer metastasis in vivo,
Page 4 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
cancer cells were injected into the perivitelline cavity of
fertilized zebrafish embryos. Four experiments were con-
ducted with ten fish per group, resulting in a total of more
than 30 zebrafish per group. e results indicated that
20% of the MDA-MB-231 cells had metastasized by day
one, an increase of 24% according to subsequent observa-
tions. In comparison, golgin-97 KO (2–9)cells presented
a greater metastasis rate, beginning at 46% on day one,
peaking at 52% on day two, and subsequently declining
to 36% on day three. Golgin-97 KO (2–20) cells pre-
sented lower rates of metastasis, with rates of 17%, 30%,
and 34%, respectively. In the present study, we present
the quantitative results obtained on day two, as shown in
Fig.1B. CM-DiI staining further revealed that the num-
ber of cancer cell clusters spread distal to the primary site
was significantly greater in fish injected with golgin-97
KO cells than in those injected with MDA-MB-231 cells
(Fig.1A and B). Furthermore, when MDA-MB-231 and
Fig. 1 Golgin-97 KO modulates the malignancy of breast cancer cells in vivo. A Representative images showing that MDA-MB-231 and golgin-97
KO cells were injected into the perivitelline cavity of zebrafish embryos and analyzed with a dissecting microscope. The location of the cancer cells
is indicated in red on bright field images of zebrafish. The white arrows indicate dispersed breast cancer cells. B Quantification of the number of cell
clusters on the basis of the acquired images in Fig. 1A. Each dot represents one live fish for image analysis. The quantitative data were analyzed
via one-way ANOVA. C Representative images of lung metastases formed from MDA-MB-231 and 2–9 KO cell-derived NOD/SCID mice on day
28 after tail vein injection. D Representative IHC images with H&E staining of lung metastases. Scale bars, 100 μm. The yellow arrows delineate
the tumor region. E Representative images showing the tumor sizes on day 42 after the injection of MDA-MB-231 and 2–9 KO cells into the right
middle mammary fat pads of the mice. F Tumor growth was monitored at the indicated times after the injection of MDA-MB-231 and 2–9 KO cells,
and tumor growth curves were constructed. The data were analyzed via two-way ANOVA. G Compared with those injected with MDA-MB-231
cells, NOD/SCID mice injected with golgin-97 KO cells exhibited poor survival (n = 9 mice per group). Kaplan–Meier plots of mouse survival
using a standard tumor volume of 300 mm3 as a criterion for euthanasia. P values were determined via the log-rank test. The data are presented
as the means ± SDs (*p < 0.05; **p < 0.01; ****, P < 0.0001)
Page 5 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
golgin-97 KO cells were injected into the lateral tail veins
of immunodeficient NOD/SCID mice, a trend toward the
formation of lung foci was observed in golgin-97 KO cell-
injected mice, suggesting that golgin-97 KO breast can-
cer cells have a greater rate of survival and colonization
success (Fig.1C). H&E staining revealed that golgin-97
KO increased the tumor cell density and resulted in the
formation of large nodules in the stroma (Fig.1D). us,
these differences in cell morphology and organization
suggest that golgin-97 KO enhances the invasive poten-
tial of cancer. In addition, the orthotopic mouse model
demonstrated that the tumors derived from golgin-97
KO cells were significantly larger than the tumors derived
from MDA-MB-231 cells (Fig.1E). Quantitative analysis
revealed a significant increase in tumor size in golgin-97
KO cell-injected mice on day 35 (Fig.1F). As expected,
the survival time of golgin-97 KO mice was shorter than
that of MDA-MB-231 mice (Fig.1G). ese results col-
lectively support the suppressive role of golgin-97 in the
tumor growth and metastatic progression of breast can-
cer invivo.
Multi‑omics reveals theinvolvement ofkey factors
ingolgin‑97 KO‑mediated breast cancer progression
To identify signaling pathways containing potential ther-
apeutic targets regulated by golgin-97, NGS and quan-
titative proteomic analysis were applied. Analysis of the
intersection of the two resulting datasets revealed that
783 DEGs were significantly upregulated, with a twofold
change in expression, in two golgin-97 KO cell lines (2–9
KO and 2–20 KO) compared with that in MDA-MB-231
cells (Fig. 2A). e IPA results suggested that several
upstream regulators and pathway networks, including
cytokines (IL-1β, TNF), growth factors (BMP4, AGT),
signal transduction factors (p38 MAPK), and transcrip-
tion factors (NF-κB-RelA), are significantly involved in
the upregulation of these 783 DEGs (Fig.2B and Table1).
e RTqPCR results verified that the expression lev-
els of three DEGs (IL-1β, IL-6, and MMP1) regulated
by the abovementioned key regulators were significantly
higher in golgin-97 KO cells than in MDA-MB-231 cells
(Fig. 2C). Additionally, DAVID bioinformatic analysis
revealed that the DEGs upregulated by golgin-97 KO
mediate biological processes such as the Wnt signaling
pathway, the MAPK cascade, wound healing, and the cel-
lular response to IL-1 (Fig.2D). We also performed TMT-
based quantitative proteomics to identify 7271 quantified
proteins in golgin-97 KO and MDA-MB-231 cells. Using
1.5 SDs of the fold change as a cutoff value, we identi-
fied 219 proteins upregulated (1.44-fold increase) by gol-
gin-97 KO. Consistent with the NGS analysis results, our
combined proteomic analysis/IPA revealed that TGFβ1,
EHF, TNF, JUNB, and IL-1α were the main activators
upregulated in response to golgin-97 KO (Fig. 2E and
Table S3). ese results were consistent with our ear-
lier study in which we utilized a quantitative proteomics
approach to establish a dataset that profiles the secreted
proteome, termed the secretome, which comprises 1,872
identified and quantified proteins from the conditioned
media of MDA-MB-231 cells, both with and without
golgin-97 knockdown. Among the proteins identified,
103 exhibited a 1.5-fold change in secretion levels. Of
these proteins, 88, including IL-6, IL-8, GM-CSF, TL1A,
PLAU, and ISG15, exhibited upregulation, whereas 15
proteins demonstrated downregulation in the golgin-
97-knockdown cells in comparison to the control cells.
Gene Ontology analysis revealed that these 103 proteins
were associated primarily with processes associated with
the wound response, cell migration, cellular localiza-
tion, cellular motility, and immune system functions (Fig.
S2). Our previous study also revealed that conditioned
medium from golgin-97-knockdown cells promoted
recipient cell migration and invasion, whereas an NF-κB
inhibitor reduced golgin-97-knockdown-induced TNBC
cell motility [17]. Taken together, these results suggest
that golgin-97 KO promotes cancer progression primar-
ily by modulating inflammatory responses and Wnt/
MAPK signaling in breast cancer cells.
Golgin‑97 KO induces tumor progression andmetastasis
accompanied bylung colonization andinammatory cell
inltration
In this study, we measured higher levels of murine TGFβ
and the proinflammatory cytokine TNFα in primary
tumors derived from golgin-97 KO cells than in those
derived from MDA-MB-231 cells (Fig. 3A), indicat-
ing that golgin-97 KO may promote tumor growth and
metastasis by modulating the tumor microenvironment.
Moreover, IHC analysis of primary tumors revealed
increased expression of the proliferation marker Ki67 and
the macrophage/monocyte marker CD68 in golgin-97
KO tumors (Fig.3B). To investigate whether golgin-97
influences macrophage polarization in the tumor micro-
environment of breast cancer, we conducted a co-culture
experiment involving cancer cells and PMA-induced
human leukemia monocytic THP-1 cells. Our results
indicated that both parental MDA-MB-231 cells and gol-
gin-97 KO cells induced a mix of pro-inflammatory and
anti-inflammatory factors production, enabling mac-
rophages to exhibit characteristics of both M1 and M2
polarization. is finding aligns with recent studies that
highlight the complexity of macrophage polarization,
where cells can display traits of both M1 and M2 pheno-
types [21, 22]. To further examine the effect of golgin-97
on macrophage polarization and function within the
tumor microenvironment associated with breast cancer
Page 6 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
Table 1 The IPA bioinformatics analysis reveals the upstream regulators for 783 up-regulated genes induced by golgin-97 knockout
Upstream regulator Molecule type p value Target molecules in dataset
IL1B Cytokine 1.00E-08 ACKR2,ADAMTS1,ANGPTL4,APOB,ASS1,BMF,C1R,C3,CABP1,CCL11,CCL20,CCL24,CEBPB,CHS
T6,CSF3,CXCL1,CYP19A1,CYP1B1,DUSP1,EBI3,ELF3,ENG,FPR2,GDF15,HAS1,HLAA,IER3,IER5L,
IL18,IL18R1,IL1A,IL1B,IL6,IL7R,INHBA,IRAK3,ITGB3,ITPKB,LCN2,LDHA,LOXL4,MAFA,MAPT,MEF
2B,MEFV,METRNL,MMP1,MMP3,MUC2,MYLK3,NOS3,PAPPA,PLXDC2,PTGS1,PTGS2,REN,SAA2
,SCX,SERPINB2,SERPINB3,SHH,SLC1A3,SOCS1,SRGN,ST18,TAP2,TGM2,TNFRSF11B,TREM1,VS
NL1,WNT5A,ZC3H12A
BMP4 Growth factor 1.15E-08 CCL11,CDH5,CEBPB,CSF3,CYP19A1,DIO2,DLX3,DLX4,ELF3,HSD3B1,IL6,INHBA,LEF1,LEFTY1,
MMP3,PGF,PITX2,PRKCH,PRR5,SHH,SNAI1,SPINT1,TAL1,TFAP2C,TGM2,TNFRSF11B,WNT5A
TNF Cytokine 1.16E-08 ACKR2,ADAMTS7,ALB,ALDH3A1,ALOX5AP,ANGPTL4,APOA1,AQP3,ASGR1,ASS1,B4GALNT
1,BDKRB2,BIK,BMP6,C1QTNF1,C3,CABP1,CCL11,CCL20,CCL24,CCN4,CD207,CD70,CDH11
,CDH5,CDO1,CEBPB,CIB2,CMKLR1,CSF3,CXCL1,CYP19A1,CYP1B1,DUSP1,DUSP14,DUSP9,
EBI3,ECH1,ELF3,ENG,FAT2,FBXO32,FPR1,FPR2,GDF15,GPRC5B,HAND1,HAS1,HCAR3,HEPA
CAM,HLAA,IER3,IL18,IL18R1,IL1A,IL1B,IL6,IL7R,INHBA,IRAK3,ITGB3,LAMP3,LCN2,LDHA,LPL,LR
IG1,LTBP2,MAFA,MEFV,METRNL,MMP1,MMP3,MUC2,MUC20,NFATC1,NOS3,PAPPA,PCK1,PDG
FA,PGF,PLA2G4C,PLXDC2,PTGS1,PTGS2,RAPGEF5,ROBO1,RRAD,SERPINB2,SHH,SLC16A2,SLC
1A3,SNAI1,SOCS1,SOX4,SYNGR3,TAPBP,TFAP2C,TG,TGM2,TH,TNFRSF11B,TNNC1,TREM1,TRI
M63,UCN2,VIPR1,WNT1,WNT3A,WNT5A,ZC3H12A,ZNF750
P38 MAPK Group 2.72E-08 ASS1,CCL11,CD70,CDH5,CEBPB,CSF3,CXCL1,CYP19A1,DIO2,DUOX2,DUSP1,FBXO32,FGF21,I
ER3,IL1A,IL1B,IL6,INHBA,ITGB3,MAL,MMP1,MMP3,MUC2,MYBPH,NFATC1,NOS3,PCK1,PTGS2,
RRAD,SERPINB2,SNAI1,TH,TJP2,TNNC1,TREM1,WNT5A
DSCAML1 Other 5.51E-08 ATOH7,BMP6,DNER,EMX1,LHX9,MAPT,NPAS3,NRCAM,NTNG2,PAX5,PCDH17,PDGFA,PRKCH,
ROBO2,SHANK1,TUBB4A
NFkB1-RelA Complex 6.06E-08 CCL11,CCL20,CSF3,CXCL1,IL1B,IL6,LCN2,MMP1,MMP3,PTGS2,TNFRSF11B
AGT Growth factor 6.53E-08 ADGRE1,AMBP,ANGPTL8,BDKRB2,BMP6,C1QTNF1,C3,CCN4,CDH5,COL4A1,COL4A2,COL6
A2,COLEC10,CYP19A1,DIO2,DUSP1,ENG,FGA,GAA,GABBR1,GAS2,GDF15,HSD3B1,IL18,IL
18R1,IL1A,IL1B,IL6,ITGB3,ITPKB,LCN2,LGI3,LOXL4,LRP1,LRRC7,LTBP2,LTBP3,MAPT,MMP1,MMP
3,MYO7A,NOS3,PDGFA,PGF,PTGS1,PTGS2,PTPN22,REN,ROBO1,SCG2,SERPINB2,SH2D1B,SLC1
A1,SOCS1,SOST,SOX4,SPINT1,SUSD5,TH,TNFRSF11B,VTN,XIRP1
Dexa-methasone Chemical drug 9.83E-08 ABCC6,ACY3,ADAMTS1,AFP,ALB,ALOX5AP,ANGPTL4,APOA1,ATF7IP2,ATP9A,AVPR1B,BDKRB2
,BMP5,C12orf42,C3,C8A,CCL11,CCL20,CDH11,CDKN1C,CEBPB,CELF2,CLDN9,CMKLR1,COL4
A1,COL4A2,COL6A2,COL7A1,CTSE,CTSW,CXCL1,CYP19A1,CYP1B1,DIO2,DNER,DUSP1,EDA,F
AM107A,FBLN1,FBXO32,FGA,FGF21,FOXQ1,FPR2,FSD2,GABBR1,GAS2L2,GBP5,GDF15,GNGT
2,GNRH2,HCST,HLAA,HSPA6,IER3,IL18,IL18R1,IL1A,IL1B,IL2RB,IL6,IL7R,INHBA,IRAK3,ITGB3,KC
NK3,KLRC4KLRK1/KLRK1,LCN2,LDHA,LEF1,LEFTY1,LPL,LRP1,LSR,LTBP2,MATN1,MATN3,MMP
1,MMP3,MUC2,NFATC1,NOS3,NPPC,NTF3,OR51E1,OSBP2,PCK1,PDGFA,PPP1R14C,PTGFRN,P
TGS1,PTGS2,PTPN22,RAB37,RPS6KA2,RRAD,SAA2,SAA4,SCG2,SERPINB2,SHH,SLC1A3,SLC2A
5,SLC4A1,SNAI1,SOCS1,SRGN,TAP2,TGM2,TLE2,TMEM61,TNFRSF11B,TNS4,TRIM63,TRPM2,T
UBB1,UCN2,VIPR1,WNT2,WNT5A,ZC3H12A,ZIC2,ZNF750
DSCAM Other 1.21E-07 EMX1,KCNJ4,MAPT,NPAS3,NRCAM,NTF3,NTNG2,PAX5,PCDH17,PDGFA,PRKCH,ROBO2,SLC1A
1,SYNGR3,TAL1,TUBB4A
CD36 Transmembrane receptor 2.19E-07 ADGRE1,APOA1,APOB,CCL20,COL4A1,CSF3,CXCL1,IL1A,IL1B,IL6,LPL,LRP1,MMP1,MMP3,SER
PINB2,TNFRSF11B
Fig. 2 Multi-omics reveals the involvement of key factors in golgin-97 KO-mediated breast cancer progression. A The DEGs between the two
golgin-97 KO cell lines and the MDA-MB-231 cells were analyzed via next-generation sequencing (NGS). The Venn diagram shows the 783
overlapping genes upregulated by golgin-97 KO. B The graphical summary of the IPA results shows the key molecules and functions of these 783
upregulated genes in golgin-97 KO cells. C qRTPCR analysis of IL-1β, IL-6, and MMP1 mRNA levels in MDA-MB-231 and golgin-97 KO cells. The data
are shown as the means ± SDs. **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 based on one-way ANOVA. D The biological processes of the 783 genes
upregulated by golgin-97 KO were analyzed with the DAVID bioinformatics resource. E Quantitative proteomics combined with bioinformatic
analysis revealed network and disease-related functions in which the 219 proteins upregulated by golgin-97 KO were involved. The orange
octagons, squares, and ovals represent functions, cytokines, and transcriptional regulators, respectively. The orange hourglasses represent canonical
pathways. The orange dotted lines indicate activation, and the gray dotted lines indicate unpredicted effects
(See figure on next page.)
Page 7 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
Fig. 2 (See legend on previous page.)
Page 8 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
invivo, we performed an association analysis of golgin-97
gene expression (GOLGA1) with M1 and M2 mac-
rophages in BRCA-Basal (TNBC > 70%) via TIMER2.0
(http:// timer. cistr ome. org/), an integrated online data-
base that compiles cancer research and immune associa-
tion data from multiple sources. Our analysis suggested
that golgin-97 may be positively correlated with M1
macrophages, as indicated by the CIBERSORT data, but
negatively correlated with M2 macrophages, accord-
ing to findings from the TIDE data (Fig. S3). Together,
these observations suggest that the downregulation
of golgin-97 in breast cancer cells may increase M2
Fig. 3 Golgin-97 KO induces tumor progression and metastasis accompanied by lung colonization and inflammatory cell infiltration. A qRTPCR
analysis of murine IL-6, TNFα, and TGFβ mRNA levels in primary tumors from mice (n = 9 mice per group). The data are based on an unpaired t test. B
Representative images of IHC staining for Ki67 and CD68 in primary tumors. Scale bars, 100 μm. C Representative images of lung metastasis sections
with H&E staining and IHC staining with anti-Ki67 and anti-CD68 antibodies. The yellow arrows delineate the tumor region. Scale bars, 200 μm (H&E
and Ki67) and 50 μm (Ki67 and CD68). D‑F Quantification of lung metastatic nodules, Ki67-positive cells, and CD68-positive cells in the MDA-MB-231
and 2–9 KO groups (n = 4 mice per group). The quantitative data were analyzed via an unpaired t test. The data are presented as the means ± SDs
(*p < 0.05; **p < 0.01; ****, P < 0.0001)
Page 9 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
macrophage polarization through complex communi-
cation between multiple cell types in tumor microen-
vironment. Next, we examined whether golgin-97 KO
promotes metastatic tumor cell colonization. H&E stain-
ing and Ki67 and CD68 expression were evaluated in lung
sections. Quantitative analysis revealed that golgin-97
KO significantly increased the nodule number and lung
metastatic colonization, as well as the infiltration of mac-
rophages/monocytes (Fig.3C-F). ese results suggested
that golgin-97 KO significantly and positively influences
primary tumor colonization and metastasis, an effect
that involves proinflammatory factor-controlled interplay
between golgin-97 KO cells and stromal cells.
Targeting ERK/MAPK kinase attenuates golgin‑97
KO‑induced breast cancer malignancy andenhances
paclitaxel treatment ecacy
To search for new therapeutic targets to benefit patients
with malignancy and chronic inflammation induced by
low expression of golgin-97, we conducted an IPA of the
783 upregulated DEGs identified from the intersection
of NGS data derived from two golgin-97 KO cell lines.
e analysis revealed a total of 629 chemical compounds,
including both activators and inhibitors. Notably, when
the predicted activation state of inhibition was examined,
36 chemical compounds were identified and subsequently
ranked on the basis of a p value of less than 0.05, with
the top five compounds documented in Table2. Among
these, we emphasized two leading chemical inhibitors:
U0126 (MEK1/2 inhibitor) and SB203580 (p38 MAPK
inhibitor). A wound healing assay revealed that gol-
gin-97 KO cells had greater collective migration capac-
ity than did MDA-MB-231 cells. Importantly, SB203580
and U0126 alone or in combination significantly reduced
golgin-97 KO-induced cell motility (Fig.4A). RTqPCR
analysis revealed that U0126 and SB203580 significantly
reduced the mRNA expression levels of IL-1β, IL-6, and
MMP1 in golgin-97 KO cells (Fig. 4B). Gene-specific
silencing via siRNA transfection was further used to con-
firm the phenomena shown by chemical inhibitor treat-
ment. As shown in Fig.4C, the mRNA expression levels
of IL-1β, IL-6, and MMP1 were decreased by p38 MAPK
knockdown. ERK1/2 knockdown reduced the mRNA
expression of IL-1β and MMP1 but not that of IL-6.
Knockdown of p38 MAPK and ERK1/2 also significantly
inhibited the increased cell migration ability induced by
golgin-97 depletion in MDA-MB-231 and MCF-7 cells
(Fig.4D-E), and the knockdown efficacy is shown in Fig.
S4A-B. Although golgin-97 KO did not promote the pro-
liferation of breast cancer cells invitro, treatment with
U0126 alone or in combination with SB203580 reduced
the viability of golgin-97 KO cells and MDA-MB 231
cells after 48 h and 72 h, respectively (Fig.4F). Paclitaxel
(PTX) is the most commonly used chemical agent for
the clinical treatment of TNBC, and long-term chemo-
therapy causes severe side effects in patients [23, 24]. We
then tested whether treatment with U0126 or SB203580
as adjuvants with paclitaxel would benefit patients with
golgin-97 deficiency. e results of the cell viability
assay revealed that treatment with PTX or two-kinase
inhibitors (U0126 and SB203580) was cytotoxic to gol-
gin-97 KO cells (Fig.4G). Notably, combined treatment
with these three drugs (U0126 + SB203580 + PTX) sig-
nificantly resulted in a synergistic effect compared with
PTX alone or the combination of U0126 and SB203580
(Fig.4G). ese results suggest that the inhibition of both
the ERK1/2 kinase pathway and the p38 MAPK pathway
has therapeutic potential for the treatment of breast can-
cer patients with low expression of golgin-97.
Inhibition ofERK/MAPK kinase iseective inovercoming
golgin‑97 KO‑induced tumor growth andlung metastasis
We next examined the therapeutic potential of ERK1/2
kinase and p38 MAPK inhibitors in an orthotopic mouse
model. We observed that treatment with PTX alone, two-
kinase inhibitors, or a combination of the three drugs
significantly reduced the tumor growth rate and tumor
volume, with targeting of ERK1/2 kinase and p38 MAPK
enhancing the efficacy of PTX-based chemotherapy.
(Fig.5A-B). Specifically, growth inhibition and survival
protection were most pronounced in the kinase inhibi-
tor treatment groups (Fig. 5B-C). To confirm whether
Fig. 4 Targeting ERK/MAPK kinase attenuates golgin-97 KO-induced breast cancer malignancy and enhances paclitaxel treatment efficacy. A
Wound healing assay of MDA-MB-231 and 2–9 KO cells treated with vehicle (veh.) control, SB203580 (20 μM), U0126 (20 μM), or SB203580 (20
μM) + U0126 (20 μM) for 17 h (left panel) and quantification of cell migration (right panel). The quantitative data were analyzed via one-way ANOVA.
qRTPCR analysis of IL-1β, IL-6, and MMP1 mRNA levels in 2–9 KO cells treated with inhibitors (B) or gene-specific knockdown (C) as indicated.
The data were analyzed via one-way ANOVA. Wound healing assays with gene-specific knockdown of p38 MAPK and ERK1/2 as indicated were
performed on 2–9 KO cells (D) and MCF-7 cells (E). The data were analyzed via one-way ANOVA. F MDA-MB-231 and 2–9 KO cells were treated
with vehicle control, SB203580, U0126, or a combination (SB203580 + U0126) for the indicated times, followed by a cell viability assay. G 2–9 KO
cells were treated with vehicle control, SB203580 + U0126, PTX, or PTX + SB203580 + U0126 for the indicated times, followed by a cell viability assay.
The data were analyzed via two-way ANOVA. The data are presented as the means ± SDs. *p < 0.05; **p < 0.01; ***, P < 0.001; ****, P < 0.0001. Images
from each group in all the wound healing assays were acquired in three random fields of view via microscopy
(See figure on next page.)
Page 10 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
Fig. 4 (See legend on previous page.)
Page 11 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
these two kinase inhibitors modulate cancer-associated
inflammation in this mouse model, the expression of
CD68 and inflammatory mediators in primary tumors
was examined. IHC revealed that PTX (PTX alone
and three drugs), but not two-kinase inhibitor, signifi-
cantly reduced CD68 expression (Fig. 5D-E). However,
qRTPCR revealed that the administration of kinase
inhibitors alone or in combination with PTX resulted in
reduced mRNA expression of murine TNFα and TGFβ
(Fig. 5F-G). Compared with that in the vehicle group,
the expression of murine TGFβ but not TNFα was lower
in the PTX alone group (Fig.5F-G). Additionally, quan-
titative analysis of IHCs obtained from lung tissue sec-
tions revealed that treatment with two-kinase inhibitors
(two-kinase inhibitors alone or three drugs) but not PTX
alone resulted in lower levels of human Ki67 expression,
indicating a reduction in lung metastatic colonization
(Fig.5H-I). Notably, we observed significant infiltration
of CD68 and induction of cleaved caspase-3 expres-
sion in lung tissues treated with PTX alone or the three
drugs, suggesting that PTX treatment may increase the
potential for lung inflammation and lung injury (Fig.5J-
L). ese results collectively suggest that ERK1/2 and
p38 MAPK inhibitors can effectively treat golgin-97-KO-
induced breast cancer malignancy by reducing inflamma-
tion at the primary tumor site and inhibiting lung organ
metastasis.
Hypoxia mimetics andERK/MAPK signaling regulate
golgin‑97 expression inbreast cancer
Hypoxia is detected in most solid tumors and strongly
promotes tumor growth and survival through metasta-
sis [2529]. Interestingly, the activation of the hypoxia-
inducible factor HIF1α/heme oxygenase-1 signaling
pathway attenuated sepsis-induced acute lung injury and
reduced Golgi stress by increasing the expression of Golgi
structural proteins such as golgin-97 [30]. We then exam-
ined whether HIF1α influences golgin-97 and MAPK
signal transduction in breast cancer cells. We detected
HIF1α accumulation with increasing doses of CoCl2 but
decreased protein levels of golgin-97, TGN46, and IκBα
(Fig. 6A-D). Upon incubation with CoCl2, ERK1/2 and
Table 2 Potential chemical inhibitors used to suppress the 783 genes upregulated by golgin-97 knockout
Upstream regulator Molecule type Predicted
activation
state
p value Target molecules in dataset
U0126 Chemical - kinase inhibitor Inhibited 6.34E-06 ADGRE1,ANGPTL4,AQP3,BMP6,C3,CCL11,CCN4,CDH5,CDKN1C,CEBPB,COL
4A1,CSF3,CXCL1,CYP19A1,CYP1B1,DUSP1,GDF15,IER3,IL1A,IL1B,IL6,INHBA
,ITGA2B,LPL,MMP1,MMP3,MUC2,MYO7A,NFATC1,NXPH4,PTGS2,REN,SERPI
NB2,SHH,SOCS1,TAP2,TGM2,TH,TNFRSF11B,ZIC2
SB203580 Chemical - kinase inhibitor Inhibited 2.38E-05 ALOX5AP,C3,CCL11,CCL20,CD207,CEBPB,CXCL1,CYP1B1,DUOX2,DUSP1,E
BI3,FGF21,FOXQ1,FPR2,HAND1,HAS1,HCAR3,HEPACAM,IER3,IL18,IL1A,IL1
B,IL6,ITGB3,MMP1,MMP3,MUC2,NOS3,PCK1,PDGFA,PTGS2,SNAI1,TGM2,TN
FRSF11B,TREM1
PD98059 Chemical - kinase inhibitor Inhibited 4.43E-05 ALOX5AP,APOA1,BDH1,C3,CCL11,CCL20,CEBPB,COL4A1,CXCL1,CYP19A1,C
YP1B1,DIO2,DUSP1,ENG,FPR2,HAS1,IER3,IL1A,IL1B,IL6,ITGA2B,ITGB3,KLRC2
,LDHA,LPL,MMP1,MMP3,MT1F,MUC2,NOS3,PCK1,PDGFA,PTGS2,REN,SERPI
NB2,SHH,SNAI1,TH,TP73
Aspirin Chemical drug Inhibited 1.17E-04 APOA1,BMP6,CDH5,CSF3,CXCL1,ENG,GDF15,IER3,IL18,IL1A,IL1B,IL6,ITPKB,
MMP1,PTGS1,PTGS2,SOX4
AS1842856 Chemical reagent Inhibited 1.20E-04 CXCL1,FBXO32,IL6,MMP1,MMP3,TRIM63
(See figure on next page.)
Fig. 5 Inhibition of ERK/MAPK kinase is effective in overcoming golgin-97 KO-induced tumor growth and lung metastasis. A NOD/SCID mice were
subcutaneously injected with 2-9 KO cells, followed by veh. control, inhibitor (SB203580+U0126), PTX, or a combination of an inhibitor with PTX
(PTX+SB203580+U0126), and tumor growth was monitored. The tumor sizes (cm) are indicated at the bottom. B Chart showing tumor sizes
and weights from 5A. The data are based on two-way ANOVA. C Survival rates of NOD/SCID mice from 5A. Kaplan–Meier plots in which the standard
tumor size (300 mm3) was used as a criterion for euthanasia in the veh.-control and drug treatment groups. D Representative images of IHC staining
for CD68 in primary tumors. Scale bars, 50 μm. E Quantification of CD68-positive cells derived from panel 5D. FG qRTPCR analysis of murine
TNFα and TGFβ mRNA levels in primary tumors from mice. The data were analyzed via one-way ANOVA. H and J Representative images of lung
metastasis sections with H&E staining and IHC staining with anti-Ki67 and anti-CD68 antibodies. The yellow arrows delineate the lung metastasis
region of the primary tumor. Quantification of Ki67-positive cells (I) and CD68-positive cells (K) from the lower panel (5H and 5J). The data were
analyzed via one-way ANOVA. L Western blotting analysis of cleaved caspase-3 and pro-caspase-3 protein levels in the lung tissues of the mice.
Actin was used as the internal control. The quantification of protein expression is shown in the lower panel. The data are presented as the means ±
SDs (*p < 0.05; **p < 0.01; ***, P < 0.001; ****, P < 0.0001)
Page 12 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
Fig. 5 (See legend on previous page.)
Page 13 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
p38 MAPK were activated following the decrease in gol-
gin-97 protein expression (Fig. 6A, E-F). ese results
suggest that, unlike acute lung injury, hypoxia may be a
physiological condition that contributes to the down-
regulation of golgin-97 expression and the activation
of ERK/MAPK signaling in cancer cells. Interestingly,
treatment of MDA-MB-231 cells with U0126 alone or
with two inhibitors (SB203580 + U0126) for 24 h caused
a significant increase in golgin-97 protein expression in
the MDA-MB-231 cells (Fig. 6G-H). A slight increase
(approximately 20%) in golgin-97 mRNA levels was
observed in combination-treated cells, suggesting an
unexpected mechanism by which MAPK signaling regu-
lates golgin-97 expression at the protein level (Fig. S5A).
Consistent with these findings, we also observed an
increase in golgin-97 protein but not golgin-97 mRNA
expression with single knockdown of ERK1/2 and double
knockdown of ERK1/2 and p38 MAPK in MDA-MB-231
and MCF-7 cells (Fig. 6I-L and S5B-3C). ese results
support the hypothesis that a potential negative regula-
tory loop exists between ERK/MAPK signaling and gol-
gin-97 expression under hypoxic conditions; therefore,
the inhibition of ERK/MAPK may abrogate the tumor-
promoting pathways induced by golgin-97 deficiency
(Fig.6M).
Discussion
Accumulating evidence indicates that the Golgi appara-
tus is a critical regulator of tumor progression. Coiled-coil
golgins play a central role in controlling membrane traf-
ficking and act as regulators of Golgi integrity and signal
transduction [8, 10, 11, 31, 32]. For example, dysregula-
tion of Golgi-localized Cdc42 signaling by the cis-Golgi
matrix protein GM130 may promote the progression of
colon and breast cancer [33, 34]. Our recent studies with
KaplanMeier plotter (http:// kmplot. com) and Oncomine
(https:// www. oncom ine. org) data revealed that low gol-
gin-97 expression is correlated with poor survival and
increased invasiveness in breast cancer cells [17]. On
the basis of the cSurvival database (https:// tau. cmmt.
ubc. ca/ cSurv ival/)) and the UALCAN database (https://
ualcan. path. uab. edu/), we also observed that low gol-
gin-97 expression significantly worsened the progno-
sis of progression-free survival (P = 0.0058). TCGA data
revealed that golgin-97 expression is notably lower in
TNBC patients than in normal controls (P < 1 × 10–12),
luminal cancer patients (P = 1.11 × 10–16), and HER2-
positive patients (P = 7.39 × 10–4) (Fig. S6). ese results
confirm that low golgin-97 expression is a key feature of
TNBC and is linked to poor clinical outcomes. e cur-
rent study showed that golgin-97 KO can promote the
metastatic dissemination and lung colonization of TNBC
cells in vivo (in zebrafish and immunodeficient mice).
Our orthotopic mouse model studies further confirmed
that golgin-97 KO has a positive regulatory effect on pri-
mary tumor growth and distant metastasis, accompanied
by a proinflammatory tumor microenvironment with
features such as increased secretion of murine tumor-
associated macrophage-related factors in tumor tissues
(Fig. 6M). Macrophages and growth factors/cytokines
directly contribute to processes facilitating breast tumor
progression and metastasis, including tumor cell growth,
migration, invasion, and intravasation [35, 36]. To assess
whether inflammatory cytokines or growth factors mod-
ulate golgin-97 expression, we administered IL-6, IL-1β,
and TGF-β to MDA-MB-231 cells. e results demon-
strated that short-term exposure to these cytokines led
to a decrease in golgin-97 mRNA expression. Prolonged
stimulation with TGF-β alone sustained the reduc-
tion in golgin-97 mRNA levels, whereas treatment with
IL-6 and IL-1β tended to lead to recovery of golgin-97
mRNA expression. Nonetheless, golgin-97 protein levels
increased following stimulation with all three cytokines
(Fig. S7). ese findings suggest a complex regulatory
mechanism involving golgin-97 in response to inflamma-
tory cytokines. Specifically, while short-term exposure to
IL-6, IL-1β, and TGF-β reduces golgin-97 mRNA levels,
the simultaneous increase in golgin-97 protein expres-
sion indicates a potential posttranscriptional regulatory
effect. is discrepancy may reflect an adaptive cellular
response in which cells prioritize the production or sta-
bilization of golgin-97 protein despite reduced mRNA
Fig. 6 Hypoxia mimetics and ERK/MAPK signaling regulate golgin-97 expression in breast cancer. A MDA-MB-231 cells were treated with increasing
concentrations of CoCl2 for 24 h and then subjected to Western blotting analysis with phosphosite- and protein-specific antibodies, as indicated.
Actin was used as the internal control. B-F Quantification of protein expression, as shown in panel 6A. G MDA-MB-231 cells treated with kinase
inhibitors were prepared for Western blotting analysis via phosphosite- and protein-specific antibodies, as indicated. Actin was used as the internal
control. H Quantification of golgin-97 protein expression levels from panel 6G. The quantitative data were analyzed via one-way ANOVA. The
golgin-97 protein level in MDA-MB-231 cells (I) and MCF-7 cells (J) following p38 MAPK, ERK1/2 individual knockdown, or combined with double
knockdown. K, L Quantification of golgin-97 protein expression levels from panel 6I6J. The data were analyzed via one-way ANOVA. The data are
presented as the means ± SDs. *p < 0.05; **p < 0.01; ***, P < 0.001 based on one-way ANOVA. M Golgin-97 deletion promotes cancer progression,
an effect that involves hypoxia, MAPK signaling, and the tumor microenvironment. MAPK inhibitors could be used to suppress golgin-97
deregulation in TNBC. This model is based in part on an illustration from biorender.com
(See figure on next page.)
Page 14 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
Fig. 6 (See legend on previous page.)
Page 15 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
levels, possibly to support functions associated with
cellular stress and inflammation. Given that neither
the knockdown nor the KO of golgin-97 significantly
increased cell proliferation in vitro, we propose that
tumor cell–stromal cell interactions have a critical impact
during golgin-97 KO-mediated cancer progression.
Currently, drugs targeting Golgi-resident proteins for
treating various cancers and metastases have entered
clinical trial stages, demonstrating the feasibility and
potential of targeting the Golgi apparatus in cancer ther-
apy [37]. However, the challenges remain the specificity of
the target, with a significant reduction in complications.
Our omics study revealed that the Wnt signaling path-
way, MAPK kinase cascades, and inflammatory cytokines
are the key regulators that are significantly upregulated
in golgin-97 KO cells. Previous studies have reported
that individual treatment with the ERK inhibitor U0126
or the p38 MAPK inhibitor SB203580 effectively inhibits
the proliferation and invasion of breast cancer cells, but
the specific regulatory factors involved remain unclear
[3843]. During cancer progression, activated mac-
rophages can polarize into two major subsets, M1 and
M2, which promote tumorigenic capabilities through the
secretion of inflammatory cytokines, including TNFα
and TGFβ [44]. Although combined treatment with
U0126 and SB203580 did not decrease CD68 infiltra-
tion in mice derived from golgin-97 KO cells, these two
kinase inhibitors significantly reduced murine TNFα and
TGFβ expression levels in primary tumors. e current
study proposes an intriguing and promising therapeutic
strategy: administering ERK/MAPK inhibitors to main-
tain tumor-suppressing golgin-97 protein expression in
TNBC, thereby attenuating breast cancer growth, the
inflammatory response, and lung metastasis. Our study
also revealed the benefits of ERK/MAPK inhibitors in
preventing lung metastasis and lung injury. Specifically,
pulmonary drug toxicity is a cause of interstitial lung
disease, which can range from benign infiltrates to life-
threatening acute respiratory distress syndrome. Direct
dose-dependent toxicity and immune inflammation are
the main causes of lung damage and fibrosis. We showed
that PTX alone but not kinase inhibitors alone caused
CD68 infiltration into lung sections and induced lung
damage. Severe lung damage occurred when PTX was
given in combination with these two inhibitors, suggest-
ing that the drug dosage and/or other unknown signal-
ing pathways contribute to this lung damage. Future work
will focus on using mouse breast cancer animal models
to reveal the interaction between golgin-97 and specific
immune cells and optimize combination treatment strat-
egies for breast cancer.
U0126 is a MEK1/2 inhibitor that primarily targets
MEK in the MAP kinase pathway, blocking downstream
ERK1/2 signaling and impacting p38 MAPK activity [45].
Notably, low-dose, short-term use of U0126 reduces p38
MAPK phosphorylation, whereas prolonged treatment
with both SB203580 and U0126 results in increased phos-
phorylation of p38 and ERK1/2. is suggests a recipro-
cal balance between the phosphorylation of ERK and p38,
where the inhibition of one pathway leads to an increase
in the activity of the other pathway [46, 47]. For exam-
ple, treatment with high concentrations of SB203580 has
been reported to activate Raf-1 and ERK signaling [48,
49], supporting potential crosstalk between ERK and p38
MAPK signaling with SB203580 in cancer therapy. U0126
may inhibit breast cancer growth by blocking the cell
cycle, but the role of p38 in cancer remains complex and
is currently a subject of ongoing debate [45, 50]. Impor-
tantly, our findings support this complexity, aligning
with the results presented in Fig.6G and Fig.4C, which
demonstrated potential crosstalk between U0126 and
SB203580. is interaction also strengthens the idea that
the combination of these two kinase inhibitors results
in antitumor activity invivo (Fig. 5) while also helping
to explain the lack of synergistic effects observed with
U0126 and SB203580 in our invitro assays (Fig.4).
Our results demonstrated that U0126 and SB203580
individually inhibited the mRNA expression of inflam-
matory cytokines and MMP1, which was upregulated by
golgin-97 KO. e increased IL-6 mRNA levels in 2–9
KO cells after ERK knockdown, as shown in Fig. 4C,
may be due to several factors. First, there is a reciprocal
relationship between the phosphorylation of ERK and
p38 MAPK, where inhibiting one pathway may increase
the other [46, 47]. Second, p38 MAPK and IL-6 have a
reciprocal and interconnected relationship in several
pathways [51]. ird, Wnt5a activates JNK, ERK1/2, and
ERK5, contributing to IL-6 expression in preadipocytes
[52]. However, loss of ERK1/2 can stimulate ERK5 sign-
aling in epithelial cells, enhancing IL-6 expression in
the tumor microenvironment [53]. We noted increased
Wnt5a in golgin-97 KO cells (data not shown), poten-
tially increasing ERK5 activity and IL-6 mRNA levels.
erefore, the increase in IL-6 following ERK knock-
down is plausible. Compared with U0126, SB203580
alone was insufficient to reduce cell viability but more
effectively inhibited the collective migration of golgin-97
KO cells, suggesting that different MAPK signaling path-
ways may be required for golgin-97 KO-induced cell
proliferation and motility (Fig.6M). e siRNA knock-
down results suggest that p38 MAPK and ERK1/2 not
only impact golgin-97 protein expression but also modu-
late cell motility and the cytotoxic effects of chemother-
apy. However, we found that SB203580 treatment alone
did not increase golgin-97 protein expression, which is
Page 16 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
inconsistent with p38 silencing. is inconsistency may
be due to the specificity and off-target effects of kinase
inhibitors [54]. Treatment with high concentrations
of SB203580 has been reported to activate Raf-1 and
ERK signaling [48, 49], supporting potential crosstalk
between ERK and p38 MAPK signaling with SB203580
in cancer therapy. Consistently, we also observed that
combined treatment with U0126 and SB203580 had the
most promising effects on cancer progression in gol-
gin-97 KO cells.
We observed that the suppression of p38 MAPK
and ERK1/2 increased the protein but not the mRNA
expression of golgin-97. ERK reportedly promotes
tumorigenesis by regulating protein stability through
the ubiquitinproteasome pathway [55]; thus, MAPK
inhibitors may maintain the protein expression of gol-
gin-97 in breast cancer cells. Consistent with this idea,
the golgin-97, IκBα, and TGN46 protein levels were
reduced, but ERK and p38 MAPK were activated in
CoCl2-treated cells, indicating an inverse correlation
between golgin-97 expression and MAPK signaling
activity (Fig. 6M). Given that CoCl2-induced HIF1α
expression occurs mainly through ERK and p38 MAPK
activation [56, 57], we cannot exclude the possibility
that ERK and p38 MAPK signaling may be upstream
regulators of golgin-97. ese findings also support
the possibility of a feedback loop between MAPKs and
golgin-97 in the modulation of breast cancer progres-
sion under hypoxic conditions. Additionally, we found
that IL-1β levels were increased in golgin-97 KO cells.
IL-1β mainly regulates cell migration by activating p38
MAPK, and its substrate MAPKAPK2 is involved in
modulating the tumor microenvironment, thereby fur-
ther promoting the invasion of breast cancer cells [58,
59]. IL-1β-induced migration of MDA-MB-231 cells
under hypoxic conditions has also been reported [60].
By using a hypoxic chamber, we found that the protein
levels of golgin-97 were increased in MDA-MB231 cells
under short-term hypoxia treatment (15 min), whereas
golgin-97 and TGN46 were significantly decreased after
2 to 8 h of hypoxic exposure (data not shown). e cur-
rent study reports the interplay between hypoxia and
the golgin-97 and MAPK signaling pathways. However,
further studies are needed to determine whether this
mechanism is directly mediated by golgin-97 in vivo.
Nevertheless, we hypothesize that hypoxia and expo-
sure to unidentified stimulators are the physiological
conditions involved in golgin-97 downregulation, which
modulates the tumor microenvironment and promotes
cell aggressiveness invivo.
To our knowledge, our study is the first to report that
golgin-97 affects tumor growth and metastasis possibly
through communication between cancer cells and inflam-
matory stromal cells. Both the anti-inflammatory effects
and the restoration of tumor-suppressive golgin-97 expres-
sion contribute to the efficacy of U0126 and SB203580
kinase inhibitors in breast cancer therapy. Compared with
the paclitaxel regimen, these two kinase inhibitors have
the potential to prevent lung injury. Although golgin-97
does not have kinase activity or directly affects protein
stability, we propose that it may indirectly affect signaling
pathways such as the ERK1/2 and p38 MAPKpathways for
three reasons. First, golgin-97 aids in transport and organi-
zation within the TGN, helping to sort signaling molecules
and receptors [61]. By ensuring correct localization, gol-
gin-97 can alter protein activation in signaling pathways.
Second, as a tethering factor, it promotes vesicle fusion
and affects access to growth factor or cytokine receptors,
such as IL-10 [62, 63]. ird, changes in Golgi integrity,
especially in cancer cells, can disrupt the processing and
glycosylation of signaling molecules, significantly alter-
ing receptor signaling and activation of the ERK1/2 and
p38 MAPKpathways [64]. Investigations of the impact of
golgin-97 on different cancers are rare, and more evidence
is needed to explore its clinical application. Overall, this
study provides new therapeutic strategies for TNBC and
may benefit drug development.
Abbreviations
TNBC Triple-negative breast cancer
KO Knockout
PTX Paclitaxel
CoCl2 Cobalt chloride
DMSO Dimethyl sulfoxide
CCK-8 Cell Counting Kit-8
MAPK Mitogen-activated protein kinase
TNF Tumor necrosis factor
TGN Trans-Golgi network
ERK Extracellular signal-regulated kinase
MEK MAPK/extracellular signal-regulated kinase
HIF1α Hypoxia-inducible factor 1 alpha
IL-1β Interleukin 1 beta
IL-6 Interleukin 6
TGFβ Transforming growth factor beta
MMP-1 Matrix metalloproteinase-1
NF-κB Nuclear factor kappa-B
qPCR Quantitative PCR; RTPCR, reverse transcription polymerase
chain reaction
TMT Tandem mass tag
2D LCMS/MS Two-dimensional liquid chromatographytandem mass
spectrometry
NGS Next-generation sequencing
IPA Ingenuity Pathway Analysis
DEGs Differentially expressed genes
DAVID Database for Annotation, Visualization and Integrated
Discovery
WT Wild-type
SDs Standard deviations
Page 17 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
Supplementary Information
The online version contains supplementary material available at https:// doi.
org/ 10. 1186/ s12964- 024- 02010-0.
The online version contains Appendix S1, Supplementary Figures S1-S7, and
Supplementary Tables S1-S3. All the data are contained within the article and/
or the supporting information. The mass spectrometry proteomics data have
been deposited to the ProteomeXchange Consortium via the PRIDE partner
repository [65] with the dataset identifier PXD056499.
Additional file 1.
Additional file 2.
Additional file 3.
Acknowledgements
We thank Ke-Wei Lin for the technical support in our genomic analysis. We
also appreciate Professor Chia-Rui Shen for providing the essential reagents for
the co-culture of cancer cells with monocytic cells.
Authors’ contributions
Y-C Liu contributed to the design of the experiments, performed the research,
analyzed the results, wrote the manuscript draft and revised the manuscript;
T-J Lin performed the research, analyzed the results and wrote the manuscript
draft; K-Y Chong contributed to the design of the experiments and performed
the research; G-Y Chen performed the research and analyzed the results; C-Y
Kuo performed the research and analyzed the results; Y-Y Lin performed the
research and analyzed the results; C-W Chang helped establish the zebrafish
model; T-F Hsiao performed the proteomic analysis; C-L Wang provided the
methodology; Y-C Shih performed the research and contributed to the data
analysis; and C-J Yu was responsible for conceptualization, experimental
design, supervision, writing, review & editing, and funding acquisition.
Data availability
The mass spectrometry proteomics data have been deposited to the Pro-
teomeXchange Consortium via the PRIDE partner repository with the dataset
identifier PXD056499 (http:// www. ebi. ac. uk/ pride).
Declarations
Ethics approval and consent to participate
The animal protocol was reviewed and approved by the Laboratory Animal
Use Committee of Chang Gung University (CGU107-228).
Competing interests
The authors declare no competing interests.
Author details
1 Department of Cell and Molecular Biology, College of Medicine, Chang Gung
University, 259 Wen-Hwa 1 road, Guishan District, Taoyuan, Taiwan. 2 CardioVas-
cular Research Center, Tzu Chi General Hospital, Hualien City, Hualien County,
Taiwan. 3 Department of Medical Biotechnology and Laboratory Sciences,
College of Medicine, Chang Gung University, Taoyuan, Taiwan. 4 Graduate
Institute of Biomedical Sciences Division of Biotechnology, College of Medi-
cine, Chang Gung University, Taoyuan, Taiwan. 5 Hyperbaric Oxygen Medical
Research Lab, Bone and Joint Research Center, Linkou Chang Gung Memorial
Hospital, Taoyuan, Taiwan. 6 Centre for Stem Cell Research, Faculty of Medicine
and Health Sciences, Universiti Tunku Abdul Rahman, Selangor, Malaysia.
7 Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung
University, Taoyuan, Taiwan. 8 School of Medicine, College of Medicine, Chang
Gung University, Taoyuan, Taiwan. 9 Department of Thoracic Medicine, Chang
Gung Memorial Hospital, Taoyuan, Taiwan. 10 Molecular Medicine Research
Center, Chang Gung University, Taoyuan, Taiwan.
Received: 22 October 2024 Accepted: 22 December 2024
References
1. DeSantis CE, Ma J, Goding Sauer A, Newman LA, Jemal A. Breast cancer
statistics, 2017, racial disparity in mortality by state. CA Cancer J Clin.
2017;67:439–48.
2. Chavez K J, Garimella SV, Lipkowitz S. Triple negative breast cancer cell
lines: one tool in the search for better treatment of triple negative breast
cancer. Breast Dis. 2010;32:35–48.
3. Obidiro O, Battogtokh G, Akala EO. Triple Negative Breast Cancer
Treatment Options and Limitations: Future Outlook. Pharmaceutics.
2023;15:1796.
4. Bielopolsk i D, Evron E, Moreh-Rahav O, Landes M, Stemmer SM, Salamon
F. Paclitaxel-induced pneumonitis in patients with breast cancer: case
series and review of the literature. J Chemother. 2017;29:113–7.
5. Limpawittayakul P, Petchjorm S, Chueansuwan W, Boonfueang W.
Paclitaxel-induced acute fibrinous and organizing pneumonitis in early
breast cancer: A case report. Respir Med Case Rep. 2024;48: 102004.
6. Mohan N, Dalip D, Rampersad FS, Jaggernauth S. Paclitaxel-Induced
Pneumonitis in Trinidad: A Case Report. Cureus. 2022;14: e26613.
7. Makaraci P, Kim K. trans-Golgi network-bound cargo traffic. Eur J Cell Biol.
2018;97:137–49.
8. Millarte V, Farhan H. The Golgi in cell migration: regulation by signal trans-
duction and its implications for cancer cell metastasis. ScientificWorld-
Journal. 2012;2012: 498278.
9. Bajaj R, Warner AN, Fradette JF, Gibbons DL. Dance of The Golgi: Under-
standing Golgi Dynamics in Cancer Metastasis. Cells. 2022;11:1484.
10. Spano D, Colanzi A. Golgi Complex: A Signaling Hub in Cancer. Cells 2022; 11.
11. Bui S, Mejia I, Diaz B, Wang Y. Adaptation of the Golgi Apparatus in Cancer
Cell Invasion and Metastasis. Front Cell Dev Biol. 2021;9: 806482.
12. Griffith KJ, Chan EK, Lung CC, Hamel JC, Guo X, Miyachi K, Fritzler MJ.
Molecular cloning of a novel 97-kd Golgi complex autoantigen associ-
ated with Sjögren’s syndrome. Arthritis Rheum. 1997;40:1693–702.
13. Lock JG, Hammond LA, Houghton F, Gleeson PA, Stow JL. E-cadherin
transport from the trans-Golgi network in tubulovesicular carriers is
selectively regulated by golgin-97. Traffic. 2005;6:1142–56.
14. Munro S, Nichols BJ. The GRIP domain - a novel Golgi-targeting domain
found in several coiled-coil proteins. Curr Biol. 1999;9:377–80.
15. Yu CJ, Lee FJ. Multiple activities of Arl1 GTPase in the trans-Golgi network.
J Cell Sci. 2017;130:1691–9.
16. Radvanyi L, Singh-Sandhu D, Gallichan S, Lovitt C, Pedyczak A, Mallo
G, Gish K, Kwok K, Hanna W, Zubovits J, et al. The gene associated with
trichorhinophalangeal syndrome in humans is overexpressed in breast
cancer. Proc Natl Acad Sci U S A. 2005;102:11005–10.
17. Hsu RM, Zhong CY, Wang CL, Liao WC, Yang C, Lin SY, Lin JW, Cheng HY,
Li PY, Yu CJ. Golgi tethering factor golgin-97 suppresses breast cancer
cell invasiveness by modulating NF-κB activity. Cell Commun Signal.
2018;16:19.
18. Lin Y, Bai L, Chen W, Xu S. The NF-kappaB activation pathways, emerging
molecular targets for cancer prevention and therapy. Expert Opin Ther
Targets. 2010;14:45–55.
19. Veinotte CJ, Dellaire G, Berman JN. Hooking the big one: the potential
of zebrafish xenotransplantation to reform cancer drug screening in the
genomic era. Dis Model Mech. 2014;7:745–54.
20. Liao WC, Lin TJ, Liu YC, Wei YS, Chen GY, Feng HP, Chang YF, Chang HT,
Wang CL, Chi HC, et al. Nuclear accumulation of KPNA2 impacts radiore-
sistance through positive regulation of the PLSCR1-STAT1 loop in lung
adenocarcinoma. Cancer Sci. 2022;113:205–20.
21. Bardi GT, Smith MA, Hood JL. Melanoma exosomes promote mixed M1
and M2 macrophage polarization. Cytokine. 2018;105:63–72.
22. Carata E, Muci M, Mariano S, Di Giulio S, Nigro A, Romano A, Panzarini E.
Extracellular Vesicles from NSC-34 MN-like Cells Transfected with Mutant
SOD1 Modulate Inflammatory Status of Raw 264.7 Macrophages. Genes
(Basel) 2024;15.
23. Crown J, O’Leary M, Ooi WS. Docetaxel and paclitaxel in the treatment of
breast cancer: a review of clinical experience. Oncologist. 2004;9(Suppl
2):24–32.
24. Gradishar WJ. Taxanes for the treatment of metastatic breast cancer.
Breast Cancer (Auckl). 2012;6:159–71.
25. Zhang J, Ouyang F, Gao A, Zeng T, Li M, Li H, Zhou W, Gao Q, Tang X,
Zhang Q, et al. ESM1 enhances fatty acid synthesis and vascular mimicry
in ovarian cancer by utilizing the PKM2-dependent warburg effect within
the hypoxic tumor microenvironment. Mol Cancer. 2024;23:94.
Page 18 of 18
Liuetal. Cell Communication and Signaling (2025) 23:22
26. K aragiota A, Kourti M, Simos G, Mylonis I. HIF-1alpha-derived cell-pene-
trating peptides inhibit ERK-dependent activation of HIF-1 and trigger
apoptosis of cancer cells under hypoxia. Cell Mol Life Sci. 2019;76:809–25.
27. Kong R, Wei W, Man Q, Chen L, Jia Y, Zhang H, Liu Z, Cheng K, Mao C, Liu
S. Hypoxia-induced circ-CDYL-EEF1A2 transcriptional complex drives
lung metastasis of cancer stem cells from hepatocellular carcinoma.
Cancer Lett. 2023;578: 216442.
28. Chen Z, Han F, Du Y, Shi H, Zhou W. Hypoxic microenvironment in cancer:
molecular mechanisms and therapeutic interventions. Signal Transduct
Target Ther. 2023;8:70.
29. Jing X, Yang F, Shao C, Wei K, Xie M, Shen H, Shu Y. Role of hypoxia in
cancer therapy by regulating the tumor microenvironment. Mol Cancer.
2019;18:157.
30. Li X, Yu J, Gong L, Zhang Y, Dong S, Shi J, Li C, Li Y, Zhang Y, Li H. Heme
oxygenase-1(HO-1) regulates Golgi stress and attenuates endotoxin-
induced acute lung injury through hypoxia inducible factor-1alpha (HIF-
1alpha)/HO-1 signaling pathway. Free Radic Biol Med. 2021;165:243–53.
31. Lu L, Tai G, Hong W. Autoantigen Golgin-97, an effector of Arl1 GTPase,
participates in traffic from the endosome to the trans-golgi network. Mol
Biol Cell. 2004;15:4426–43.
32. Yadav S, Puri S, Linstedt AD. A primary role for Golgi positioning in
directed secretion, cell polarity, and wound healing. Mol Biol Cell.
2009;20:1728–36.
33. Baschieri F, Confalonieri S, Bertalot G, Di Fiore PP, Dietmaier W, Leist M,
Crespo P, Macara IG, Farhan H. Spatial control of Cdc42 signalling by a
GM130-RasGRF complex regulates polarity and tumorigenesis. Nat Com-
mun. 2014;5:4839.
34. Baschieri F, Uetz-von Allmen E, Legler DF, Farhan H. Loss of GM130 in
breast cancer cells and its effects on cell migration, invasion and polarity.
Cell Cycle. 2015;14:1139–47.
35. Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell
migration, invasion, and metastasis. Cell. 2006;124:263–6.
36. Tang XQ. Tumor-associated macrophages as potential diagnostic and
prognostic biomarkers in breast cancer. Cancer Lett. 2013;332:3–10.
37. Zappa F, Failli M, De Matteis MA. The Golgi complex in disease and
therapy. Curr Opin Cell Biol. 2018;50:102–16.
38. Uehara N, Matsuoka Y, Tsubura A. Mesothelin promotes anchorage-inde-
pendent growth and prevents anoikis via extracellular signal-regulated
kinase signaling pathway in human breast cancer cells. Mol Cancer Res.
2008;6:186–93.
39. Yang F, Tang XY, Liu H, Jiang ZW. Inhibition of mitogen-activated protein
kinase signaling pathway sensitizes breast cancer cells to endoplasmic
reticulum stress-induced apoptosis. Oncol Rep. 2016;35:2113–20.
40. Duzgun SA, Yerlikaya A, Zeren S, Bayhan Z, Okur E, Boyaci I. Differential
effects of p38 MAP kinase inhibitors SB203580 and SB202190 on growth
and migration of human MDA-MB-231 cancer cell line. Cytotechnology.
2017;69:711–24.
41. He Q, Xue S, Tan Y, Zhang L, Shao Q, Xing L, Li Y, Xiang T, Luo X, Ren G.
Dual inhibition of Akt and ERK signaling induces cell senescence in triple-
negative breast cancer. Cancer Lett. 2019;448:94–104.
42. Ren J, Wang Y, Ware T, Iaria J, Ten Dijke P, Zhu HJ. Reactivation of BMP sign-
aling by suboptimal concentrations of MEK inhibitor and FK506 reduces
organ-specific breast cancer metastasis. Cancer Lett. 2020;493:41–54.
43. Wang Z, Wang F, Ding XY, Li TE, Wang HY, Gao YH, Wang WJ, Liu YF, Chen
XS, Shen KW. Hippo/YAP signaling choreographs the tumor immune
microenvironment to promote triple negative breast cancer progression
via TAZ/IL-34 axis. Cancer Lett. 2022;527:174–90.
44. Zhang Q, Sioud M. Tumor-Associated Macrophage Subsets: Shaping
Polarization and Targeting. Int J Mol Sci 2023;24.
45. You Y, Niu Y, Zhang J, Huang S, Ding P, Sun F, Wang X. U0126: Not only a
MAPK kinase inhibitor. Front Pharmacol. 2022;13: 927083.
46. Hotokezaka H, Sakai E, Kanaoka K, Saito K, Matsuo K, Kitaura H, Yoshida N,
Nakayama K. U0126 and PD98059, specific inhibitors of MEK, accelerate
differentiation of RAW264.7 cells into osteoclast-like cells. J Biol Chem
2002; 277:47366–47372.
47. Poddar R, Paul S. Novel crosstalk between ERK MAPK and p38 MAPK
leads to homocysteine-NMDA receptor-mediated neuronal cell death. J
Neurochem. 2013;124:558–70.
48. Birkenkamp KU, Tuyt LM, Lummen C, Wierenga AT, Kruijer W, Vellenga E.
The p38 MAP kinase inhibitor SB203580 enhances nuclear factor-kappa B
transcriptional activity by a non-specific effect upon the ERK pathway. Br
J Pharmacol. 2000;131:99–107.
49. Feng L, Xie X, Ding Q, Luo X, He J, Fan F, Liu W, Wang Z, Chen Y. Spatial
regulation of Raf kinase signaling by RKTG. Proc Natl Acad Sci U S A.
2007;104:14348–53.
50. Low HB, Zhang Y. Regulatory Roles of MAPK Phosphatases in Cancer.
Immune Netw. 2016;16:85–98.
51. Krupkova O, Sadowska A, Kameda T, Hitzl W, Hausmann ON, Klasen J,
Wuertz-Kozak K. p38 MAPK Facilitates Crosstalk Between Endoplasmic
Reticulum Stress and IL-6 Release in the Intervertebral Disc. Front Immu-
nol. 2018;9:1706.
52. Díaz-Chamorro S, Garrido-Jiménez S, Barrera-López JF, Mateos-Quirós CM,
Cumplido-Laso G, Lorenzo MJ, Román ÁC, Bernardo E, Sabio G, Carvajal-
González JM, Centeno F. Title: p38δ Regulates IL6 Expression Modulating
ERK Phosphorylation in Preadipocytes. Front Cell Dev Biol. 2021;9: 708844.
53. de Jong PR, Taniguchi K, Harris AR, Bertin S, Takahashi N, Duong J, Cam-
pos AD, Powis G, Corr M, Karin M, Raz E. ERK5 signalling rescues intestinal
epithelial turnover and tumour cell proliferation upon ERK1/2 abrogation.
Nat Commun. 2016;7:11551.
54. Weiss WA, Taylor SS, Shokat KM. Recognizing and exploiting differ-
ences between RNAi and small-molecule inhibitors. Nat Chem Biol.
2007;3:739–44.
55. Yang JY, Zong CS, Xia W, Yamaguchi H, Ding Q, Xie X, Lang JY, Lai CC,
Chang CJ, Huang WC, et al. ERK promotes tumorigenesis by inhibiting
FOXO3a via MDM2-mediated degradation. Nat Cell Biol. 2008;10:138–48.
56. Chen R, Xu J, She Y, Jiang T, Zhou S, Shi H, Li C. Necrostatin-1 pro-
tects C2C12 myotubes from CoCl2-induced hypoxia. Int J Mol Med.
2018;41:2565–72.
57. Wang G, Li Y, Yang Z, Xu W, Yang Y, Tan X. ROS mediated EGFR/MEK/ERK/
HIF-1alpha Loop Regulates Glucose metabolism in pancreatic cancer.
Biochem Biophys Res Commun. 2018;500:873–8.
58. Gelfo V, Romaniello D, Mazzeschi M, Sgarzi M, Grilli G, Morselli A, Manzan
B, Rihawi K, Lauriola M. Roles of IL-1 in Cancer: From Tumor Progression to
Resistance to Targeted Therapies. Int J Mol Sci. 2020;21:6009.
59. Rebe C, Ghiringhelli F. Interleukin-1beta and Cancer. Cancers (Basel).
2020;12:1791.
60. Filippi I, Carraro F, Naldini A. Interleukin-1β Affects MDAMB231 Breast
Cancer Cell Migration under Hypoxia: Role of HIF-1α and NFκB Transcrip-
tion Factors. Mediators Inflamm. 2015;2015: 789414.
61. Shin JJH, Crook OM, Borgeaud AC, Cattin-Ortolá J, Peak-Chew SY, Breckels
LM, Gillingham AK, Chadwick J, Lilley KS, Munro S. Spatial proteomics
defines the content of trafficking vesicles captured by golgin tethers. Nat
Commun. 2020;11:5987.>
62. Stanley AC, Lieu ZZ, Wall AA, Venturato J, Khromykh T, Hamilton NA,
Gleeson PA, Stow JL. Recycling endosome-dependent and -independent
mechanisms for IL-10 secretion in LPS-activated macrophages. J Leukoc
Biol. 2012;92:1227–39.
63. Murray RZ, Stow JL. Cytokine Secretion in Macrophages: SNAREs, Rabs,
and Membrane Trafficking. Front Immunol. 2014;5:538.
64. Li Y, Mu L, Li Y, Mi Y, Hu Y, Li X, Tao D, Qin J. Golgi dispersal in cancer stem
cells promotes chemoresistance of colorectal cancer via the Golgi stress
response. Cell Death Dis. 2024;15:417.
65. Griss J, Perez-Riverol Y, Lewis S, Tabb DL, Dianes JA, Del-Toro N, Rurik M,
Walzer MW, Kohlbacher O, Hermjakob H, et al. Recognizing millions of
consistently unidentified spectra across hundreds of shotgun proteomics
datasets. Nat Methods. 2016;13:651–6.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.
... The MAPK pathway, consisting of ERK1/2, MEK1/2 and p38 (38), is one of the most commonly mutated oncogenic pathways in several cancers (39), including colorectal cancer, lung cancer, and thyroid cancer. The abnormal activation of proteins such as EGFR, RAS (including KRAS, HRAS, NRAS), and RAF (such as BRAF) can lead to MAPK pathway dysregulation, resulting in uncontrolled cellular proliferation and dedifferentiation (40). ...
Article
Full-text available
Colorectal cancer (CRC) is one of the most prevalent and life-threatening malignancies worldwide. Jujuboside B (JUB) is a bioactive compound derived from the seeds of Ziziphus jujuba, known for its potential anticancer properties. The present study aimed to investigate the association of JUB with inhibiting the proliferation, apoptosis and ferroptosis of human CRC cells with mitogen-activated protein kinase (MAPK) pathway regulation. First, the human CRC HCT116 cell line was treated with different concentrations of JUB. Subsequently, cell viability was evaluated using MTT assay and colony formation was assessed using a colony formation assay. Flow cytometry was used to detect cell apoptosis and the levels of reactive oxygen species. Western blotting was utilized to assess the expression levels of apoptosis-related proteins, ferroptosis regulators and MAPK pathway-related proteins. In addition, biochemical assay kits were used to evaluate the levels of malondialdehyde, glutathione, total iron and ferrous iron. The results demonstrated that cell viability and colony formation were markedly decreased after JUB treatment, whilst the level of apoptosis was notably increased in a concentration-dependent manner. Using electron microscopy, cells treated with JUB exhibited typical apoptotic bodies, as well as mitochondrial swelling and cristae disruption, further demonstrating JUB-induced cell apoptosis. Western blot analysis indicated that JUB treatment markedly reduced the expression of B-cell lymphoma-2 (Bcl-2) but notably increased the expression of Bcl-2 associated X-protein and cleaved caspase-3. Additionally, JUB induced ferroptosis and inhibited the MAPK signaling pathway in CRC cells. Collectively, the findings of the present study suggest that JUB has the potential to inhibit CRC cell proliferation and induce apoptosis through regulating the MAPK pathway. Therefore, JUB may be a promising therapeutic agent for the treatment of CRC.
Article
Full-text available
Chemotherapy is a crucial treatment for colorectal tumors. However, its efficacy is restricted by chemoresistance. Recently, Golgi dispersal has been suggested to be a potential response to chemotherapy, particularly to drugs that induce DNA damage. However, the underlying mechanisms by which Golgi dispersal enhances the capacity to resist DNA-damaging agents remain unclear. Here, we demonstrated that DNA-damaging agents triggered Golgi dispersal in colorectal cancer (CRC), and cancer stem cells (CSCs) possessed a greater degree of Golgi dispersal compared with differentiated cancer cells (non-CSCs). We further revealed that Golgi dispersal conferred resistance against the lethal effects of DNA-damaging agents. Momentously, Golgi dispersal activated the Golgi stress response via the PKCα/GSK3α/TFE3 axis, resulting in enhanced protein and vesicle trafficking, which facilitated drug efflux through ABCG2. Identification of Golgi dispersal indicated an unexpected pathway regulating chemoresistance in CRC.
Article
Full-text available
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease targeting the brain and spinal cord. Non-neuronal cells, including macrophages, may contribute to the disruption of motor neurons (MNs), neuromuscular junction dismantling and clinical signs of ALS. Understanding the modality and the effect of MNs–macrophage communication is pivotal. Here, we focus on extracellular vesicle (EVS)-mediated communication and, in particular, we analyze the response of macrophages. NSC-34 cells transfected with mutant SOD1 (G93A, A4V, G85R, G37R) and differentiated towards MN-like cells, and Raw 264.7 macrophages are the cellular models of the study. mSOD1 NSC-34 cells release a high number of vesicles, both large-lEVs (300 nm diameter) and small-sEVs (90 nm diameter), containing inflammation-modulating molecules, and are efficiently taken up by macrophages. RT-PCR analysis of inflammation mediators demonstrated that the conditioned medium of mSOD1 NSC-34 cells polarizes Raw 264.7 macrophages towards both pro-inflammatory and anti-inflammatory phenotypes. sEVs act on macrophages in a time-dependent manner: an anti-inflammatory response mediated by TGFβ firstly starts (12 h); successively, the response shifts towards a pro-inflammation IL-1β-mediated (48 h). The response of macrophages is strictly dependent on the SOD1 mutation type. The results suggest that EVs impact physiological and behavioral macrophage processes and are of potential relevance to MN degeneration.
Article
Full-text available
Background The hypoxic tumor microenvironment is a key factor that promotes metabolic reprogramming and vascular mimicry (VM) in ovarian cancer (OC) patients. ESM1, a secreted protein, plays an important role in promoting proliferation and angiogenesis in OC. However, the role of ESM1 in metabolic reprogramming and VM in the hypoxic microenvironment in OC patients has not been determined. Methods Liquid chromatography coupled with tandem MS was used to analyze CAOV3 and OV90 cells. Interactions between ESM1, PKM2, UBA2, and SUMO1 were detected by GST pull-down, Co-IP, and molecular docking. The effects of the ESM1-PKM2 axis on cell glucose metabolism were analyzed based on an ECAR experiment. The biological effects of the signaling axis on OC cells were detected by tubule formation, transwell assay, RT‒PCR, Western blot, immunofluorescence, and in vivo xenograft tumor experiments. Results Our findings demonstrated that hypoxia induces the upregulation of ESM1 expression through the transcription of HIF-1α. ESM1 serves as a crucial mediator of the interaction between PKM2 and UBA2, facilitating the SUMOylation of PKM2 and the subsequent formation of PKM2 dimers. This process promotes the Warburg effect and facilitates the nuclear translocation of PKM2, ultimately leading to the phosphorylation of STAT3. These molecular events contribute to the promotion of ovarian cancer glycolysis and vasculogenic mimicry. Furthermore, our study revealed that Shikonin effectively inhibits the molecular interaction between ESM1 and PKM2, consequently preventing the formation of PKM2 dimers and thereby inhibiting ovarian cancer glycolysis, fatty acid synthesis and vasculogenic mimicry. Conclusion Our findings demonstrated that hypoxia increases ESM1 expression through the transcriptional regulation of HIF-1α to induce dimerization via PKM2 SUMOylation, which promotes the OC Warburg effect and VM.
Article
Full-text available
Background Paclitaxel is a chemotherapeutic drug widely used in breast cancer treatment. While common side effects are possible, paclitaxel-induced pneumonitis is rare, with an estimated incidence of 1%–5% and a high mortality rate. Case presentation A 57-year-old Thai woman diagnosed with stage II right breast cancer. She received adjuvant chemotherapy comprising doxorubicin and cyclophosphamide, followed by weekly paclitaxel. After the ninth paclitaxel cycle, she developed acute respiratory failure. Transbronchial biopsies revealed acute fibrinous and organizing pneumonitis. The patient was placed in prone position. Following the administration of dexamethasone, her symptoms improved. However, while reducing the dexamethasone dosage, she developed new-onset dyspnea as well as Takotsubo cardiomyopathy. Intravenous methylprednisolone 500 mg/day was administered for 3 days followed by transition to intravenous dexamethasone and slow tapering to prednisolone. Prednisolone was gradually tapered and eventually discontinued after 3 months. Conclusions Paclitaxel-induced pneumonitis is a rare complication. The diagnosis should be considered in any patient who develops respiratory symptoms while receiving paclitaxel. Acute fibrinous and organizing pneumonitis is a rare type of interstitial pneumonitis with high recurrence and mortality rates. High-dose steroids are needed to treat this type of pneumonitis.
Article
Full-text available
Triple negative breast cancer (TNBC) has a negative expression of estrogen receptors (ER), progesterone receptors (PR), and human epidermal growth factor receptors (HER2). The survival rate for TNBC is generally worse than other breast cancer subtypes. TNBC treatment has made significant advances, but certain limitations remain. Treatment for TNBC can be challenging since the disease has various molecular subtypes. A variety of treatment options are available, such as chemotherapy, immunotherapy, radiotherapy, and surgery. Chemotherapy is the most common of these options. TNBC is generally treated with systemic chemotherapy using drugs such as anthracyclines and taxanes in neoadjuvant or adjuvant settings. Developing resistance to anticancer drugs and off-target toxicity are the primary hindrances to chemotherapeutic solutions for cancer. It is imperative that researchers, clinicians, and pharmaceutical companies work together to develop effective treatment options for TNBC. Several studies have suggested nanotechnology as a potential solution to the problem of suboptimal TNBC treatment. In this review, we summarized possible treatment options for TNBC, including chemotherapy, immunotherapy, targeted therapy, combination therapy, and nanoparticle-based therapy, and some solutions for the treatment of TNBC in the future. Moreover, we gave general information about TNBC in terms of its characteristics and aggressiveness.
Article
Full-text available
The tumor microenvironment (TME) is a critical regulator of tumor growth, progression, and metastasis. Among the innate immune cells recruited to the tumor site, macrophages are the most abundant cell population and are present at all stages of tumor progression. They undergo M1/M2 polarization in response to signals derived from TME. M1 macrophages suppress tumor growth, while their M2 counterparts exert pro-tumoral effects by promoting tumor growth, angiogenesis, metastasis, and resistance to current therapies. Several subsets of the M2 phenotype have been observed, often denoted as M2a, M2b, M2c, and M2d. These are induced by different stimuli and differ in phenotypes as well as functions. In this review, we discuss the key features of each M2 subset, their implications in cancers, and highlight the strategies that are being developed to harness TAMs for cancer treatment.
Article
Full-text available
Having a hypoxic microenvironment is a common and salient feature of most solid tumors. Hypoxia has a profound effect on the biological behavior and malignant phenotype of cancer cells, mediates the effects of cancer chemotherapy, radiotherapy, and immunotherapy through complex mechanisms, and is closely associated with poor prognosis in various cancer patients. Accumulating studies have demonstrated that through normalization of the tumor vasculature, nanoparticle carriers and biocarriers can effectively increase the oxygen concentration in the tumor microenvironment, improve drug delivery and the efficacy of radiotherapy. They also increase infiltration of innate and adaptive anti-tumor immune cells to enhance the efficacy of immunotherapy. Furthermore, drugs targeting key genes associated with hypoxia, including hypoxia tracers, hypoxia-activated prodrugs, and drugs targeting hypoxia-inducible factors and downstream targets, can be used for visualization and quantitative analysis of tumor hypoxia and antitumor activity. However, the relationship between hypoxia and cancer is an area of research that requires further exploration. Here, we investigated the potential factors in the development of hypoxia in cancer, changes in signaling pathways that occur in cancer cells to adapt to hypoxic environments, the mechanisms of hypoxia-induced cancer immune tolerance, chemotherapeutic tolerance, and enhanced radiation tolerance, as well as the insights and applications of hypoxia in cancer therapy.
Article
Full-text available
U0126, as an inhibitor of the MAPK signaling pathway, is closely related to various biological processes, such as differentiation, cell growth, autophagy, apoptosis, and stress responses. It makes U0126 play an essential role in balancing cellular homeostasis. Although U0126 has been suggested to inhibit various cancers, its complete mechanisms have not been clarified in cancers. This review summarized the most recent and relevant research on the many applications of U0126 and described its role and mechanisms in different cancer cell types. Moreover, some acknowledged functions of U0126 researched in the laboratory were listed in our review. We discussed the probability of using U0126 to restain cancers or suppress the MAPK pathway as a novel way of cancer treatment.
Article
Full-text available
Paclitaxel-induced pneumonitis (PIP) is an immune-mediated disease resulting from a delayed hypersensitivity reaction (type IV) to paclitaxel, an anti-microtubule chemotherapeutic drug commonly used to treat breast cancer in both neoadjuvant and adjuvant settings. PIP is diagnosed by exclusion utilizing laboratory work-up, imaging, biopsy studies, and results of antibiotic therapy because there is no single diagnostic test. Ground-glass opacifications on CT, coupled with minimal restrictive disturbance with decreased diffusion on pulmonary function tests (PFTs), negative bronchoalveolar lavage (BAL), and bronchoscopy cultures, may assist physicians in diagnosing paclitaxel-induced pneumonitis. In this report, we describe a case of PIP present in Trinidad, West Indies, which has not been described previously in this region.
Article
Hepatocellular carcinoma (HCC) is often associated with poor outcomes due to lung metastasis. ICAM-1+ circulating tumor cells, termed circulating cancer stem cells (CCSCs), possess stem cell–like characteristics. However, it is still unexplored how their presence indicates lung metastasis tendency, and particularly, what mechanism drives their lung metastasis. Here, we demonstrated that a preoperative CCSC count in 5 mL of blood (CCSC5) of >3 was a risk factor for lung metastasis in clinical HCC patients. The CSCs overexpressed with circ-CDYL entered the bloodstream and developed lung metastases in mice. Mechanistically, circ-CDYL promoted COL14A1 expression and thus ERK signaling to facilitate epithelial-mesenchymal transition. Furthermore, we uncovered that an RNA-binding protein, EEF1A2, acted as a novel transcriptional (co-) factor to cooperate with circ-CDYL and initiate COL14A1 transcription. A high circ-CDYL level is caused by HIF-1⍺-mediated transcriptional upregulation of its parental gene CDYL and splicing factor EIF4A3 under a hypoxia microenvironment. Hence, the hypoxia microenvironment enables the high-tendency lung metastasis of ICAM-1+ CCSCs through the HIF-1⍺/circ-CDYL-EEF1A2/COL14A1 axis, potentially allowing clinicians to preoperatively detect ICAM-1+ CCSCs as a real-time biomarker for precisely deciding HCC treatment strategies.