Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2020

Open Access 01.12.2020 | Research

BDH2 triggers ROS-induced cell death and autophagy by promoting Nrf2 ubiquitination in gastric cancer

verfasst von: Jia-Zhou Liu, Yi-Lin Hu, Ying Feng, Yun Jiang, Yi-Bing Guo, Yi-Fei Liu, Xi Chen, Jun-Ling Yang, Yu-yan Chen, Qin-Sheng Mao, Wan-Jiang Xue

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2020

Abstract

Background

3-Hydroxy butyrate dehydrogenase 2 (BDH2) is a short-chain dehydrogenase/reductase family member that plays a key role in the development and pathogenesis of human cancers. However, the role of BDH2 in gastric cancer (GC) remains largely unclear. Our study aimed to ascertain the regulatory mechanisms of BDH2 in GC, which could be used to develop new therapeutic strategies.

Methods

Western blotting, immunohistochemistry, and RT-PCR were used to investigate the expression of BDH2 in GC specimens and cell lines. Its correlation with the clinicopathological characteristics and prognosis of GC patients was analysed. Functional assays, such as CCK-8 and TUNEL assays, transmission electron microscopy, and an in vivo tumour growth assay, were performed to examine the proliferation, apoptosis, and autophagy of GC cells. Related molecular mechanisms were clarified by luciferase reporter, coimmunoprecipitation, and ubiquitination assays.

Results

BDH2 was markedly downregulated in GC tissues and cells, and the low expression of BDH2 was associated with poor survival of GC patients. Functionally, BDH2 overexpression significantly induced apoptosis and autophagy in vitro and in vivo. Mechanistically, BDH2 promoted Keap1 interaction with Nrf2 to increase the ubiquitination level of Nrf2. Ubiquitination/degradation of Nrf2 inhibited the activity of ARE to increase accumulation of reactive oxygen species (ROS), thereby inhibiting the phosphorylation levels of AktSer473 and mTORSer2448.

Conclusions

Our study indicates that BDH2 is an important tumour suppressor in GC. BDH2 regulates intracellular ROS levels to mediate the PI3K/Akt/mTOR pathway through Keap1/Nrf2/ARE signalling, thereby inhibiting the growth of GC.
Begleitmaterial
Additional file 1 Table S1. Sequences of primers used for amplification of target genes. Fig. S1 Inhibition of autophagy inhibits BDH2-induced apoptosis. A In BDH2-overexpressing SGC7901 and BGC823 cells, the expression levels of LC3-II, p62, cleaved caspase-3, and PARP were determined by western blotting after treatment with the autophagy inhibitor 3-MA. B In BDH2-overexpressing SGC7901 and BGC823 cells, cell viability was measured by CCK8 assays in the absence or presence of 3-MA. C In BDH2-overexpressing SGC7901 and BGC823 cells, apoptosis was assessed by flow cytometry in the absence or presence of 3-MA. *p < 0.05, **p < 0.01. Fig. S2 BDH2 overexpression triggers ROS generation. Left panel: Detection of intracellular ROS levels by fluorescence microscopy (magnification, × 200, scale bars, 20 μm). Right panel: Quantitative representation of ROS production indicated by fluorescence signal intensities. *p < 0.05; ** p < 0.01. Fig. S3 Effect of BDH2 on Keap1 and Nrf2 mRNA levels. The mRNA levels of Keap1 and Nrf2 were measured by qRT-PCR. Results are presented as means ± S.D. (n = 3); ns, not significant. Fig. S4 BDH2-induced ROS have an important role in the PI3K/Akt/mTOR pathway. A Levels of relevant signalling pathway proteins in BDH2-overexpressing SGC7901 and BGC823 cells were examined by western blotting. B Protein expression levels of p-AktSer473 and p-mTORSer2448 were detected in the presence or absence of NAC by western blotting. Fig. S5 Effect of BDH2 on intracellular iron levels. Cells expressing BDH2 or vector were analyzed for intracellular iron concentration by colorimetry. Results are presented as means ± S.D. (n = 3); ns, not significant.
Hinweise
Jia-Zhou Liu, Yi-Lin Hu and Ying Feng contributed equally to this work.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13046-020-01620-z.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
GC
Gastric cancer
BDH2
3-Hydroxy butyrate dehydrogenase 2
Baf-A1
Bafilomycin A1
3-MA
3-methyladenine
ARE
Antioxidant response elements
Nrf2
Nuclear factor erythroid 2-related factor 2
HO-1
Heme oxygenase 1
NQO1
NAD(P)H–quinone oxidoreductase 1
ROS
Reactive oxygen species
Keap1
Kelch-like ECH-associated protein 1
NAC
N-acetylcysteine

Background

Gastric cancer (GC) is one of the most common malignant tumours in the world with morbidity and mortality accounting for the fourth and second places among malignant tumours. Each year, more than 800,000 new patients are diagnosed with GC, of which nearly 90% have advanced GC, and few patients are eligible for surgery. Because of the heterogeneity of GC, the efficacy of traditional radiotherapies and chemotherapies is not satisfactory. In recent years, biotherapy and targeted therapy for GC have made great progress, but the prognosis of patients with GC is still not optimistic, and the molecular mechanisms of GC occurrence and development are still unclear [1]. Autophagy is a common physiological process in normal and GC cells. Abnormal levels of autophagy have major effects on the occurrence and progression of GC. Therefore, elucidating the mechanism of autophagy in the development of GC has great clinical significance.
Reactive oxygen species (ROS) are important signalling molecules in cells, which participate in the transmission of information via multiple signalling pathways [2, 3]. Excessive ROS induce tumour cell autophagy and apoptosis by inhibiting PI3K/Akt and other pathways, thereby inhibiting the occurrence and development of tumours [4]. For example, salinomycin promotes autophagy and apoptosis of prostate cancer cells through PI3K/Akt/mTOR and ERK/p38 MAPK pathways by increasing the cellular ROS level [5]. Inhibiting the autophagy level of prostate cancer cells increases their apoptosis level induced by salinomycin, thereby increasing the chemotherapy sensitivity of salinomycin. Ciclopirox olamine increases ROS levels in rectal cancer cells by affecting mitochondrial functions and then induces apoptosis and protective autophagy through the AMPK pathway [6]. Inhibiting this cytoprotective autophagy increases the level of apoptosis in rectal cancer cells induced by ciclopirox olamine. ROS in tumour cells are not only produced by stimulation from the external environment, but are also generated by the cell itself or as a byproduct of other biological reactions [79]. Post-translational modifications of proteins, such as ubiquitination and phosphorylation, play an important role in this process. For example, changes in the level of nuclear factor erythroid 2-related factor 2 (Nrf2) SUMOylation affect the intracellular ROS level and thus the autophagy level of cells, which ultimately affect the occurrence and development of hepatocellular carcinoma [10].
BDH2 is a short-chain dehydrogenase/reductase family member originally named as DHRS6 [11]. The human BDH2 gene is located on 4q and encodes 245 amino acids. BDH2 is widely expressed in the cytoplasm of epithelial cells of organs such as the kidney, small intestines, oral cavity, and breast. BDH2 may be a multifunctional gene in mammalian cells. It is a novel cytosolic-type 2-hydroxybutyrate dehydrogenase and has a physiological role in the use of cytosolic ketone bodies that can subsequently enter mitochondria and the tricarboxylic acid cycle [12]. BDH2 also catalyses the synthesis of 2,5-dihydroxybenzoic acid during biosynthesis of enterobactin [13]. In addition, BDH2 may be an independent poor prognosis marker of acute myeloid leukaemia by affecting apoptosis. BDH2 is regulated by long-chain non-coding RNA TP73-AS1, which affects oesophageal squamous cell carcinoma cell proliferation and apoptosis [14]. However, the effect of BDH2 expression on autophagy of GC cells and its possible mechanism have not been reported.
In this study, we analysed the expression level of BDH2 in paired GC tissues by immunohistochemical analysis, and the correlation between BDH2 expression levels and clinicopathological features and prognosis of GC. We found that low expression of BDH2 is an independent molecular marker of a poor prognosis of GC patients. GC cell lines stably expressing BDH2 were constructed, and the effect of the change of the BDH2 expression level on the apoptosis and autophagy of GC cells was observed. On the molecular level, we demonstrated that BDH2 regulates the Keap1-Nrf2 interaction and promotes proteasomal degradation of Nrf2. Increasing the levels of ubiquitination/degradation of Nrf2 led to intracellular ROS accumulation, thereby altering the PI3K/Akt /mTOR pathway to inhibit the growth of GC cells in vivo and in vitro.

Materials and methods

Patients and tissue samples

We collected 171 paired tumour and adjacent non-cancerous gastric tissues from January 2010 to December 2010 at the Affiliated Hospital of Nantong University (Jiangsu, China). Detailed clinicopathological information is provided in Table 1. Another cohort of 30 fresh GC cases was collected from the Department of General Surgery for western blot analysis. All patients were definitively diagnosed with GC by pathological histology and had not received adjuvant chemotherapy, radiation therapy, or immunotherapy before surgical excision of tumours. All experiments were conducted with the approval of the Committee for the Ethical Review of Research involving Human Subjects at Nantong University Affiliated Hospital. Written informed consent was obtained from each patient before sample collection.
Table 1
Correlation between BDH2 expression in GC tissues and clinicopathological features of GC patients
Clinicopathological parameter
N
BDH2 expression
p value
Low 127
High 44
Gender
   
0.371
 Male
111
80(72.1%)
31(27.9%)
 
 Female
60
47(78.3%)
13(21.7%)
 
Age (years)
 
.
.
0.433
 ≤60
73
52(71.2%)
21(28.8%)
 
 > 60
98
75(76.5%)
23(23.5%)
 
Degree of differentiation
   
0.177
 Well
18
11(61.1%)
7(38.9%)
 
 Moderate/Poor
153
116(75.8%)
37(24.2%)
 
Tumor diameter (cm)
   
0.124
 < 5
112
79(70.5%)
33(29.5%)
 
 ≥5
59
48(81.4%)
11(18.6%)
 
Tumor localization
   
0.536
 Up
19
13(68.4%)
6(31.6%)
 
 Middle/Down
152
114(75.0%)
38(25.0%)
 
TNM stage
   
0.021
 I + II
95
64(67.3%)
31(32.7%)
 
 III
76
63(82.9%)
13(17.1%)
 
Depth of invasion
   
0.008
 T1 + T2
72
46(63.8%)
26(36.2%)
 
 T3 + T4
99
81(81.8%)
18(18.2%)
 
Lymph node metastasis
   
0.047
 Negative
79
53(67.1%)
26(32.9%)
 
 Positive
92
74(80.4%)
18(19.6%)
 

Cell culture and reagents

Seven GC cell lines (AGS, BGC823, MGC803, MKN45, MKN1, SGC7901, and HGC27), a gastric mucosa cell line (GES-1), and human kidney cell line (HEK293T) were purchased from GeneChem (Shanghai, China). Cells were cultured in RPMI-1640 medium supplemented with 10% FBS at 37 °C in a humidified atmosphere with 5% CO2.
DAPI, Baf-A1, and 3-MA were obtained from Solarbio (Beijing, China). NAC was purchased from Beyotime (Shanghai, China), and 740Y-P (also known as 740YPDGFR) was obtained from MCE (Monmouth Junction, NJ, USA).

Plasmids and cell transfection

FLAG-tagged BDH2, HA-tagged Keap1, and MYC-tagged Nrf2 overexpression plasmid were purchased from GeneChem (Shanghai, China). To construct the BDH2 expression vector, the full-length open reading frame of human BDH2 cDNA was cloned into the eukaryotic expression vector pcDNA3.1 (Invitrogen). Expression vectors were transfected into cells with low expression of BDH2 (SGC7901 and BGC823) using Lipofectamine 3000 (Invitrogen). Stably transfected cells were selected for more than 2 weeks using neomycin (G418; Roche, Indianapolis, IN, USA).

Cell proliferation, clonogenic assay, cell cycle analysis, and apoptosis assay

These experiments were performed as described previously [15].

Immunofluorescence staining

Immunofluorescence staining was performed as described in our previous study [15]. The cells were incubated with anti-LC3B (1:1000, Abcam, ab48394) and anti-Nrf2 (1:200, Proteintech, 16,396) antibodies at 4 °C overnight. After washing three times with PBS, the cells were stained with secondary antibodies (ABclonal) at 37 °C for 2 h. Finally, Nuclei were labelled with 0.1 g/mL DAPI for 15 min, except when visualising autophagosomes (LC3-II) using an LSM780 laser-scanning confocal microscope (Carl Zeiss). Other images were captured under a BX41 fluorescence microscope (Olympus, Japan).

Transmission electron microscopy (TEM)

As described previously [15], transfected cells and xenografted tumour tissues were fixed in glutaraldehyde, and then ultrathin sections were prepared with an EM UC7 ultramicrotome (Leica, Solms, Germany). The sections were then imaged by a transmission electron microscope (Hitachi Scientific Instruments, Mountain View, CA, USA) to detect autophagic vacuoles.

Immunohistochemistry (IHC)

The IHC protocol was performed as described previously [15]. Briefly, tumour sections of patients and nude mouse xenografts were analysed by IHC using the EnVision™ System (Dako, Carpinteria, CA). Primary antibodies used for IHC were anti-BDH2 (1:100, Proteintech, 27,207), anti-Ki67 (1:100, Abcam, ab15580), anti-cleaved caspase-3 (1:200, CST, #9661), anti-LC3B (1:200, Abcam, ab48394), anti-Nrf2 (1:50, Proteintech, 16,396), anti-p-Akt (1:100, Proteintech, 66,444), and anti-p-mTOR (1:100, Abcam, ab109268).

In vivo tumourigenesis assays

Male BALB/c nude mice were randomly divided into two groups (n = 6/group). Two groups of mice were subcutaneously injected with 1 × 107 cells transfected with an empty vector or BDH2 expression vector into the right side of the neck. After tumours were visible, the tumour size was measured every 3 days until 30 days. The formula volume = (length×width^2)/2 cm2 was used to calculate tumour volumes. The mice were sacrificed after 30 days, and the tumour weight was measured. The xenografted tumours were fixed for histological analysis. A TUNEL assay was performed using an In Situ Cell Death Detection Kit (Roche; Mannheim, Germany). The Animal Care Committee of Nantong University reviewed and approved all animal experiments.

Reactive oxygen species (ROS) assay

Measurement of intracellular ROS was performed using 2,7-dichlorofluorescin-diacetate (DCFH-DA) (Yeasen, Shanghai, China). Cells were incubated with 10 μM DCFH-DA diluted in RPMI-1640 medium for 30 min at 37 °C in the dark. The cells were then immediately observed under the fluorescence microscope. Fluorescence intensity was quantified using ImageJ software. For flow cytometry, cells were collected for analysis. For tissue ROS measurement, frozen tumour sections were incubated with DCFH-DA (Item No: E004, Jiancheng Bioengineering, Nanjing, China) at 37 °C for 30 min. The ROS level in the tissue was measured under the BX41 fluorescence microscope.

Autophagic flux analysis

As described previously [15], cells were transfected with GFP-mRFP-LC3 (Shanghai, China) for 24 h and then treated for 48 h with or without 10 nM Baf-A1. Images were obtained under a confocal microscope (Carl Zeiss).

Quantitative real-time PCR (qRT-PCR)

Total RNA in GC cells was extracted with Trizol reagent (Invitrogen). qRT-PCR was carried out as described previously [16]. Primer sequences are listed in Table S1.

Transient transfection and luciferase assay

A pGL3-ARE-Luc reporter was obtained from Genechem, which is a ready-to-transduce ARE responsive lentiviral firefly luciferase reporter to monitor the transcriptional activity of Nrf2. Cells were cultured in 6-well plates and then transfected with the pGL3-ARE-Luc reporter plasmid and pRL-TK plasmid (Promega) using Lipofectamine 3000. A Luciferase Reporter Gene Assay Kit (Beyotime Institute of Biotechnology) was used to determine ARE-driven promoter activity. Luciferase activity is expressed as the ratio to that in control cells.

Coimmunoprecipitation (co-IP)

Total cell lysates were incubated with 1 μg primary antibody or negative control rabbit IgG at 4 °C overnight. Then, 20 μl protein A + G agarose (Bioworld Technology, St. Louis Park, MN, USA) was added, followed by further incubation for 2 h at 4 °C. Protein–antibody complexes were rinsed four times with PBS, and the beads were collected by centrifugation. Proteins were detected by SDS-polyacrylamide gel electrophoresis and western blotting.

Western blotting

Total protein separation and western blotting were performed as described previously [16]. Western blot analysis was performed using anti-cleaved PARP (#5625), anti-cleaved caspase-3 (#9661), anti-p-AMPK (Thr172) (#50081), anti-p-ERK1/2 (#4370), and anti-HA-Tag (#3724) antibodies purchased from Cell Signaling Technology (Danvers, MA, USA). Anti-LC3 (14600–1-AP), anti-P62/SQSTM1 (18420–1-AP), anti-phospho-Akt (Ser-473) (66444–1-Ig), anti-Akt (10176–2-AP), anti-mTOR (20657–1-AP), anti-P38 MAPK (14064–1-AP), anti-JNK (51151–1-AP), anti-ERK1/2 (16443–1-AP), anti-AMPK (10929–2-AP), anti-Nrf2 (16396–1-AP), anti-Keap1 (10503–2-AP), anti-FLAG-tag (66008–2-AP), anti-MYC-tag (60003–2-AP), and anti-β-actin (66009–1-Ig) antibodies were obtained from Proteintech Group Co., Ltd. (Wuhan, China). An anti-phospho-mTOR (Ser-2448) (ab109268) antibody was purchased from Abcam (Cambridge, MA, USA). Anti-p-p38 (sc-7973), anti-p-JNK (sc-6254), and anti-Ub (sc-8017) antibodies were obtained from Santa Cruz Biotechnology., Inc. (Santa Cruz, CA, USA).

Ubiquitination assay

For the polyubiquitinated Nrf2 assay, whole cell lysates prepared with RIPA buffer containing a proteinase inhibitor were subjected to IP of endogenous Nrf2 protein. The levels of Nrf2 ubiquitination were detected by immunoblotting with an anti-Ub antibody.

Assessment of intracellular Iron levels

Iron levels were measured by Iron Colorimetric Assay Kit (ABIN411680, Biovision) according to the manufacturer’s instructions [13].

Statistical analysis

Data are presented as means ± standard deviation (SD). All experiments were performed at least three times. Statistical analysis was performed with the SPSS software version 22 and GraphPad Prism 7.0 software. The survival rates patients with GC were calculated by Kaplan-Meier and log-rank analyses, and prognostic factors were evaluated using the Cox regression model. Student’s t-test was used for statistical comparisons between experimental groups. p < 0.05 was considered as statistically significant.

Results

BDH2 is downregulated in GC tissues

To investigate the role of BDH2 in GC, we first analysed the BDH2 mRNA level in the Oncomine database (https://​www.​oncomine.​org) and found that its expression was significantly lower in GC tissues compared with corresponding normal tissues (Fig. 1a). We also observed a similar down-regulation of BDH2 expression in GC in the UALCAN dataset (http://​ualcan.​path.​uab.​edu/​index.​html) (Fig. 1b). We next analysed BDH2 protein expression in matched gastric normal (N) and cancerous (T) tissues by western blotting. As shown in Fig. 1c, d, lower BDH2 expression was observed in eight of 10 GC cases compared with matched adjacent normal tissues (p < 0.05). Immunohistochemical analysis was subsequently performed on a TMA of 171 GC and matched adjacent normal samples. Low protein expression of BDH2 was observed in 74.3% (127/171) of GC tissues compared with matched adjacent non-cancerous tissue samples (p < 0.01; Fig. 1e and f). These results suggest that BDH2 expression is downregulated in GC.
We then analysed the BDH2 expression status together with various clinicopathological features of the 171 GC patients (Table 1). The results showed that low expression of BDH2 was positively associated with the TNM stage, depth of invasion, and lymph node metastasis, but not with other clinicopathological features including gender, age, differentiation, tumour localization, or tumour size. Moreover, multiple logistic regression analysis showed that low expression of BDH2 was significantly correlated with depth of invasion (p < 0.05, odds ratio [OR] = 0.461, 95% confidence interval [CI]: 0.214–0.993).
In addition, Kaplan-Meier analysis showed that GC patients with low BDH2 expression had worse OS and DFS rates than those with high BDH2 expression (Fig. 1g). Moreover, Cox multivariate analysis showed that low expression of BDH2 was an independent predictor of OS and DFS (Table 2).
Table 2
Univariate and multivariable analysis of OS and DFS of patients with GC
Variable
OS
DFS
Univariate analysis
P > |z|
Multivariable analysis
Univariate analysis
P > |z|
Multivariable analysis
P > |z|
HR (95% CI)
P > |z|
HR (95% CI)
Gender
 Male (n = 111) vs. female (n = 60)
0.290
  
0.264
  
Age (years)
  ≤ 60 (n = 73) vs.
> 60 (n = 98)
0.787
  
0.805
  
Differentiation
 Well (n = 18) vs. Moderate/Poor (n = 153)
0.075
  
0.073
  
Tumor diameter (cm)
  < 5 (n = 112) vs. ≥5 (59)
0.015
  
0.019
  
Tumor localization
 Up (n = 19) vs. Middle/Down (152)
0.425
  
0.429
  
Depth of invasion
 T1 + T2 (n = 72) vs. T3 + T4 (n = 99)
< 0.001
0.003
2.431(1.346-4.390)
< 0.001
0.004
2.367(1.311-4.274)
Lymph node metastasis
 Negative (n = 79) vs. Positive (n = 92)
< 0.001
0.001
2.642(1.513-4.615)
< 0.001
0.001
2.653(1.518-4.637)
BDH2 expression
 Low (n = 127) vs. High (n = 44)
0.001
0.011
0.404(0.200-0.815)
0.001
0.017
0.426(0.211-0.861)

Ectopic expression of BDH2 inhibits GC via inhibiting cell proliferation and inducing cell cycle arrest and apoptosis

Real-time PCR and western blot analyses were performed to investigate the mRNA and protein levels of BDH2 in seven GC cell lines and GES-1 cells. The BDH2 mRNA and protein levels in GC cell lines were significantly lower than those in GES-1 cells (Fig. 2a and b). To elucidate the biological functions of BDH2 in GC, SGC7901 and BGC823 cells with the lowest endogenous levels of BDH2 were stably transfected with the BDH2 expression vector. Re-expression of BDH2 protein in these cells was confirmed by western blotting (Fig. 2c). Overexpression of BDH2 significantly inhibited cell proliferation as evidenced by CCK-8 and clonogenic assays of SGC7901 and BGC823 cells (Fig. 2d and e).
To examine the molecular mechanism of BDH2-mediated inhibition of cell growth, we determined the effects of BDH2 on cell cycle progression and apoptosis by flow cytometry. The results showed that BDH2 overexpression resulted in significant cell cycle arrest at G0/G1 phase and induced apoptosis (Fig. 2f and g). To confirm the effects of BDH2 on apoptosis, apoptosis markers were analysed by western blotting. The results indicated that expression of cleaved caspases-3 and cleaved PARP was significantly increased in BDH2-overexpressing cells (Fig. 2h). Taken together, these results indicated that overexpression of BDH2 significantly inhibited the growth of GC cells.

BDH2 inhibits tumour growth in vivo

To investigate the effect of BDH2 on GC growth in vivo, a nude mouse xenograft model was established by subcutaneous injection of BDH2- or empty vector-transfected SGC7901 cells. As shown in Fig. 3a-c, the volume and net weight of tumours expressing BDH2 were significantly smaller compared with the empty vector group. Moreover, immunohistochemical analysis of the xenografts showed that BDH2 overexpression reduced the expression of Ki-67 and enhanced the expression of cleaved caspase-3 (Fig. 3d). TUNEL analysis also indicated that the apoptosis rate was increased in the BDH2-expressing group (Fig. 3e). These results demonstrated that BDH2 overexpression inhibits GC growth in vivo.

BDH2 induces autophagy in GC cells

Autophagy is a type of programmed cell death in addition to apoptosis. To examine whether the cytotoxic effect of BDH2 resulted from autophagy activation, we detected the expression levels of two autophagy-related proteins by western blotting. As shown in Fig. 4a, BDH2 overexpression potently increased the protein level of LC3-II, but downregulated the protein expression level of p62. In addition, TEM showed that formation of autophagic vacuoles was frequent in BDH2-overexpressing cells (Fig. 4b). Furthermore, endogenous LC3 puncta were remarkably increased in BDH2-overexpressing GC cells (Fig. 4c). After GFP-mRFP-LC3 lentivirus transfection, the numbers of red puncta (autophagolysosomes) were increased in BDH2-overexpressing SGC7901 and BGC823 cells, indicating an increase in autophagic flux. When bafilomycin A1 (Baf-A1) pretreatment was performed to block autophagic flux, more yellow dots (autophagosomes) were observed (Fig. 4d-g). Furthermore, we investigated the effect of BDH2 overexpression on GC autophagy in vivo. IHC staining indicated that the expression level of LC3B was upregulated in xenografts of BDH2-overexpressing cells (Fig. 4h). Moreover, TEM results demonstrated that BDH2 overexpression significantly increased autophagic double-membrane compartments containing lamellar structures (Fig. 4i). These data indicated that BDH2 overexpression induced autophagy in GC cells.

BDH2 induces cell death by promoting autophagosome formation in GC cells

There is increasing evidence of a complex link between autophagy and apoptosis. To analyse whether there was an intrinsic link between BDH2-induced GC apoptosis and autophagy, we pretreated SGC7901 and BGC823 cells with 3-methyladenine (3-MA; an autophagy inhibitor) that prevents the formation of autophagosomes. The results showed that 3-MA treatment not only inhibited BDH2 overexpression-induced autophagy, but also decreased BDH2-induced apoptosis caused by active caspase-3 and cleaved PARP (Fig. S1A). The viability of BDH2-overexpressing cells was decreased significantly after treatment with 3-MA (Fig. S1B). In addition, 3-MA treatment inhibited overexpressed BDH2-induced GC cell apoptosis (Fig. S1C). These data revealed that autophagy induced by BDH2 had a pro-death function in GC cells.

BDH2 induces apoptosis and autophagy via ROS in GC cells

Previous studies have demonstrated that ROS are responsible for triggering cell death and autophagy in tumours [1719]. Therefore, we analysed the effects of BDH2 overexpression on ROS levels in GC cells. Flow cytometry indicated that BDH2 overexpression increased ROS levels in GC cells (Fig. 5a and b). Fluorescence microscopy also showed that BDH2 overexpression increased the DCF fluorescence intensity in GC cells (Fig. S2). To examine whether there was an intrinsic link between ROS accumulation caused by BDH2 overexpression and the changes in biological behaviours of GC cells, we pretreated GC cells with the ROS inhibitor N-acetylcysteine (NAC). The pretreatment with NAC inhibited apoptosis (Fig. 5c and d) and autophagy (Fig. 5e-h) in BDH2-overexpressing cells. Moreover, western blot assays showed that pretreatment with NAC reversed the change in expression levels of cleaved caspase-3, cleaved PARP, LC3-II, and p62 in BDH2-overexpressing cells (Fig. 5i). Interestingly, we found that tissue samples derived from the BDH2-treated group showed stronger DCF fluorescence (Fig. 5j). Collectively, these findings revealed that BDH2 induces cellular ROS accumulation, thereby promoting apoptosis and autophagic cell death.

BDH2 increases the intracellular ROS level in GC cells by enhanced ubiquitination and degradation of Nrf2

It has been reported that the kelch-like ECH-associated protein 1 (Keap1)-Nrf2-antioxidant response elements (ARE) signal transduction pathway maintains intracellular redox homeostasis by regulating ROS levels [20]. To investigate the underlying mechanism of ROS accumulation caused by BDH2, we examined the effect of BDH2 on expression of Nrf2 target genes heme oxygenase 1 (HO-1) and NAD(P)H-quinone oxidoreductase 1 (NQO1). qRT-PCR analyses showed that the transcription levels of HO-1 and NQO1 were markedly downregulated in BDH2-overexpressing cells (Fig. 6a). Furthermore, a luciferase reporter plasmid containing an ARE sequence was transfected into HEK293T, SGC7901, and BGC823 cells, and luciferase assays indicated that BDH2 overexpression reduced the activity of the ARE in the cells (Fig. 6b). To reveal the cause of ARE promoter activation, we analysed the expression levels of Keap1 and Nrf2 in BDH2-overexpressing cells by qRT-PCR and western blot. BDH2 did not change the mRNA level of Nrf2 or Keap1 (Fig. S3). However, BDH2 significantly decreased the protein level of Nrf2. Conversely, the protein level of Keap1 was not affected (Fig. 6c). Moreover, immunofluorescence revealed that the level of Nrf2 was significantly decreased in BDH2-overexpressing cells (Fig. 6d). Collectively, these results suggested that BDH2 overexpression downregulates Nrf2 expression through its protein stability rather than transcription.
To gain insight into the molecular mechanistic basis of BDH2 regulating the Keap1-Nrf2 pathway, we overexpressed BDH2 plasmid with Flag-tag, HA-tagged Keap1 and Myc-tagged Nrf2 in HEK293T cell line. After immunoprecipitation using antibody against Flag-tag, HA-tagged Keap1 and Myc-tagged Nrf2 were not detected with anti-HA and anti-Myc antibody, indicating BDH2 failed to interact with Keap1 or Nrf2 (Fig. 6e). Interestingly, Co-IP showed that overexpressing BDH2 increased the interaction of endogenous Nrf2 with Keap1 (Fig. 6f). Previous research has shown that Keap1 mediates proteasomal degradation of Nrf2 by recruiting E3 ubiquitin ligase Cullin 3. To examine whether BDH2 decreased the level of Nrf2 by affecting ubiquitination/degradation of Nrf2, the ubiquitination level of Nrf2 was investigated in BDH2-overexpressing cells. As a result, we found that BDH2 overexpression promoted the ubiquitination of Nrf2 (Fig. 6g). A similar tendency was consistently observed by IHC staining of Nrf2. Nrf2-positive cells were also decreased in BDH2-expressing xenografts (Fig. 6h). These results revealed that BDH2 reduced the Nrf2 protein level by inhibiting its dissociation from E3 ubiquitin ligase Keap1, thereby reducing activity of the ARE promoter and correspondingly downregulating the transcription levels of HO-1 and NQO1.

ROS/PI3K/Akt/mTOR axis contributes to BDH2-induced apoptosis and autophagy initiation

To further explore the underlying mechanism of BDH2-induced apoptosis and autophagy, we examined the effect of BDH2 on the expression of classic autophagy-related pathways such as AMPK, MAPK/Erk1/2, and PI3K/Akt/mTOR signalling [21, 22]. As shown in Fig. 7a and Fig. S4A, compared with the empty vector group, no significant changes in the expression of AMPK or MAPK/Erk1/2 were found in BDH2-overexpressing cells. However, the expression levels of p-AktSer473 and p-mTORSer2448 were significantly lower than in the vector group (Fig. 7a; Fig. S4A). Pretreatment with 740Y-P (PI3K agonist) inhibited downregulation of p-AktSer473 and p-mTORSer2448 expression induced by overexpression of BDH2 (Fig. 7b). Furthermore, the 740Y-P pretreatment downregulated the expression levels of cleaved caspase-3, cleaved PARP, and LC3-II and upregulated the expression level of p62 in GC cells expressing BDH2 (Fig. 7c). Consistent with these results, BDH2-induced apoptosis and autophagy were decreased after 740Y-P pretreatment (Fig. 7d-i). Accordingly, IHC revealed that BDH2 overexpression downregulated the expression of p-AktSer473 and p-mTORSer2448 in xenografted tumours (Fig. 7j). These results showed that BDH2 enhanced apoptosis and autophagy of GC cells via blocking PI3K/Akt/mTOR signalling.
Next, we elucidated the effect of ROS accumulation on the PI3K/Akt/mTOR pathway. As shown in Fig. S4B, pretreatment with NAC reversed the change in expression levels of p-AktSer473 and p-mTORSer2448 in BDH2-overexpressing cells. In summary, these data indicated that BDH2 inhibits the activity of the PI3K/Akt/mTOR pathway by inducing accumulation of ROS in GC cells.

Discussion

In this study, we found that BDH2 was significantly down-regulated in GC tissues and that BDH2 expression correlated with adverse clinicopathological parameters in GC. Furthermore, Kaplan–Meier analysis showed that GC patients with low expression of BDH2 had a poor prognosis. Hypermethylation of the gene promoter region is one of the important mechanisms of gene inactivation. We used MethPrimer (http://​www.​urogene.​org/​methprimer/​) and did not find any CpG island in the BDH2 promoter region, indicating that the down-regulation or deletion of the BDH2 gene in GC tissues may not be related to DNA promoter methylation. The occurrence of GC is the result of a combination of multiple factors. For example, long-term chronic stimulation of various inflammatory factors is a key factor leading to the occurrence of GC [23, 24]. Previous studies have shown that sustained inflammation can cause down-regulation of BDH2 expression [25]. Therefore, we speculate that long-term continuous stimulation of inflammatory factors during the development of GC is an important cause of down-regulation of BDH2 expression in GC tissues.
In the present study, we identified that overexpression of BDH2 inhibited GC cell growth in vitro and in vivo. Iron participates in various biological functions, such as cell proliferation, growth, and ferroptosis [26]. Evidence from recent studies showed that the mammalian siderophore is an important regulator of cellular iron homeostasis [27]. Notably, previous studies have shown that biosynthesis of the mammalian siderophore (2,5-DHBA) is catalyzed by BDH2 [27]. However, whether the biological function induced by BDH2 is related to intracellular iron levels remains unclear. Here, we used a colorimetric assay to measure cytoplasmic iron concentration in GC cells. Our results indicated that overexpression of BDH2 had little effect on iron levels in GC cells (Fig. S5).
AMPK, MAPK/Erk1/2, and PI3K/Akt/mTOR signalling pathways are classic apoptosis and autophagy pathways [2830]. Previous studies have shown that BDH2 inhibits HCC cell growth, proliferation, and migration by inducing apoptosis and autophagy [31]. However, the specific molecular mechanisms of BDH2-induced apoptosis and autophagy remained unclear. Thus, we explored whether BDH2 participates in the regulation of three classic pathways in GC cells. Western blotting indicated that the phosphorylation levels of AktSer473 and mTORSer2448 were downregulated in BDH2-overexpressing cells. However, no significant changes in the expression of p-AMPK, p-p38 MAPK, p-JNK, or p-ERK were found in BDH2-overexpressing cells. Additionally, expression of p-AktSer473 and p-mTORSer2448 was reversed after pretreatment with 740Y-P. Interestingly, pretreatment with 740Y-P also significantly reversed the formation of autophagic flux and enhancement of LC3-II. Moreover, 740Y-P pretreatment reduced BDH2-induced cell death. These results indicated that BDH2 suppressed the growth of GC cells via inhibition of PI3K/Akt/mTOR signalling.
ROS play a key role in the induction of apoptosis and autophagy [32]. ROS are important signalling molecules in cells, which participate in multiple signalling pathways. A recent report revealed that ROS participate in the induction of caspase-independent cell death in macrophages [33, 34]. Moreover, ROS induce autophagy and apoptosis of tumour cells by inhibiting pathways such as PI3K/Akt and JNK, thereby suppressing the malignant phenotype of tumour cells [35, 36]. In this study, a significant increase in ROS generation was detected by flow cytometry and DCFH-DA in the BDH2-overexpressing group, which was clearly inhibited by a ROS scavenger (NAC). We found that BDH2 overexpression triggered apoptosis and autophagy in GC cells by inducing ROS production. However, treatment with NAC drastically rescued apoptotic cell death. In addition, NAC pretreatment remarkably inhibited autophagic flux and conversion of LC3-I to LC3-II. Furthermore, NAC reversed the expression levels of p-AktSer473 and p-mTORSer2448 in BDH2-overexpressing cell lines. Taken together, these results implied that BDH2-induced apoptosis and autophagy were triggered through ROS-mediated PI3K/Akt/mTOR pathways.
ROS produced by oxidative stress are regulated by Nrf2 and its downstream target genes including HO-1 and NQO1 [3740]. In healthy cells, Nrf2 binds to Keapl in the cytoplasm, which then recruits E3 ubiquitin ligase Cullin 3, thereby facilitating rapid degradation of Nrf2 by proteasomes [4143]. However, under oxidative stress conditions, Keap1 is uncoupled from Nrf2, allowing Nrf2 to transfer into the nucleus and bind to the ARE, thereby activating downstream antioxidants to increase cellular antioxidant activity. Nrf2 is highly expressed in all kinds of tumour tissues including GC, and cancer cells employ the cytoprotective action of Nrf2 to counter a microenvironment that is not conducive for tumour growth [4447]. Several mechanisms have been reported for continuous activation of Nrf2 in GC. A nonsynonymous somatic mutation (G333C) was detected in the first repeat of the double glycine repeat/Kelch domain, which does not repress the activity of Nrf2 [48]. The increased expression of Nrf2 helps to maintain ROS levels below a toxic threshold to escape death in cancer cells [49]. Recent studies have shown that inhibition of Nrf2 activity suppresses tumour growth and enhances the therapeutic efficiency of chemotherapeutic drugs against cancer [5053]. Therefore, under different pathological conditions, analysing Nrf2 sheds light on cancer prevention and treatment. In the present study, we found that BDH2 overexpression inhibited ARE-mediated antioxidant gene expression. Moreover, BDH2 downregulated Nrf2 expression through its protein stability rather than transcription. Co-IP verified the effect of BDH2 on binding of Keap1 and Nrf2. Our results suggested that BDH2 stabilises the binding of Keap1 and Nrf2 to promote ubiquitination/degradation of Nrf2 in GC cells.

Conclusions

In conclusion, we have identified a novel tumour suppressor gene, BDH2, which is frequently downregulated in GC. BDH2-induced ROS accumulation suppresses phosphorylation of AktSer473 and mTORSer2448 by promoting ubiquitination of Nrf2, thereby inhibiting the growth of GC cells and playing a tumour-suppressive role (Fig. 8). These findings indicate that BDH2 may be a potential new target for the treatment of GC.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s13046-020-01620-z.

Acknowledgements

The authors sincerely thank Jing-jing Lu for her guildance and suggestions for this study.

Competing interest

The authors declare no conflicts of interest.
In animal experiments, we comply with the ARRIVE guidelines, the UK Animals (Scientific Procedures) Act, 1986 and associated guidelines, EU Directive 2010/63/EU for animal experiments, or the National Institutes of Health guide for the care and use of Laboratory animals (NIH Publications No. 8023, revised 1978). The mice purchased are male. In addition, animal experiments were performed using protocols approved by the Animal Center of the Medical College of Nantong University.
Not applicable.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary information

Additional file 1 Table S1. Sequences of primers used for amplification of target genes. Fig. S1 Inhibition of autophagy inhibits BDH2-induced apoptosis. A In BDH2-overexpressing SGC7901 and BGC823 cells, the expression levels of LC3-II, p62, cleaved caspase-3, and PARP were determined by western blotting after treatment with the autophagy inhibitor 3-MA. B In BDH2-overexpressing SGC7901 and BGC823 cells, cell viability was measured by CCK8 assays in the absence or presence of 3-MA. C In BDH2-overexpressing SGC7901 and BGC823 cells, apoptosis was assessed by flow cytometry in the absence or presence of 3-MA. *p < 0.05, **p < 0.01. Fig. S2 BDH2 overexpression triggers ROS generation. Left panel: Detection of intracellular ROS levels by fluorescence microscopy (magnification, × 200, scale bars, 20 μm). Right panel: Quantitative representation of ROS production indicated by fluorescence signal intensities. *p < 0.05; ** p < 0.01. Fig. S3 Effect of BDH2 on Keap1 and Nrf2 mRNA levels. The mRNA levels of Keap1 and Nrf2 were measured by qRT-PCR. Results are presented as means ± S.D. (n = 3); ns, not significant. Fig. S4 BDH2-induced ROS have an important role in the PI3K/Akt/mTOR pathway. A Levels of relevant signalling pathway proteins in BDH2-overexpressing SGC7901 and BGC823 cells were examined by western blotting. B Protein expression levels of p-AktSer473 and p-mTORSer2448 were detected in the presence or absence of NAC by western blotting. Fig. S5 Effect of BDH2 on intracellular iron levels. Cells expressing BDH2 or vector were analyzed for intracellular iron concentration by colorimetry. Results are presented as means ± S.D. (n = 3); ns, not significant.
Literatur
1.
Zurück zum Zitat Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomark Prev. 2014;23:700–13. Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomark Prev. 2014;23:700–13.
2.
Zurück zum Zitat Kaushik S, Shyam H, Agarwal S, Sharma R, Nag TC, Dwivedi AK, et al. Genistein potentiates Centchroman induced antineoplasticity in breast cancer via PI3K/Akt deactivation and ROS dependent induction of apoptosis. Life Sci. 2019;117073. Kaushik S, Shyam H, Agarwal S, Sharma R, Nag TC, Dwivedi AK, et al. Genistein potentiates Centchroman induced antineoplasticity in breast cancer via PI3K/Akt deactivation and ROS dependent induction of apoptosis. Life Sci. 2019;117073.
3.
Zurück zum Zitat Wang L, Wang C, Tao Z, Zhao L, Zhu Z, Wu W, et al. Curcumin derivative WZ35 inhibits tumor cell growth via ROS-YAP-JNK signaling pathway in breast cancer. J Exp Clin Cancer Res. 2019;38:460.PubMedPubMedCentral Wang L, Wang C, Tao Z, Zhao L, Zhu Z, Wu W, et al. Curcumin derivative WZ35 inhibits tumor cell growth via ROS-YAP-JNK signaling pathway in breast cancer. J Exp Clin Cancer Res. 2019;38:460.PubMedPubMedCentral
4.
Zurück zum Zitat Wen C, Wang H, Wu X, He L, Zhou Q, Wang F, et al. ROS-mediated inactivation of the PI3K/AKT pathway is involved in the antigastric cancer effects of thioredoxin reductase-1 inhibitor chaetocin. Cell Death Dis. 2019;10:809.PubMedPubMedCentral Wen C, Wang H, Wu X, He L, Zhou Q, Wang F, et al. ROS-mediated inactivation of the PI3K/AKT pathway is involved in the antigastric cancer effects of thioredoxin reductase-1 inhibitor chaetocin. Cell Death Dis. 2019;10:809.PubMedPubMedCentral
5.
Zurück zum Zitat Kim KY, Park KI, Kim SH, Yu SN, Park SG, Kim YW, et al. Inhibition of autophagy promotes Salinomycin-induced apoptosis via reactive oxygen species-mediated PI3K/AKT/mTOR and ERK/p38 MAPK-dependent signaling in human prostate Cancer cells. Int J Mol Sci. 2017;18. Kim KY, Park KI, Kim SH, Yu SN, Park SG, Kim YW, et al. Inhibition of autophagy promotes Salinomycin-induced apoptosis via reactive oxygen species-mediated PI3K/AKT/mTOR and ERK/p38 MAPK-dependent signaling in human prostate Cancer cells. Int J Mol Sci. 2017;18.
6.
Zurück zum Zitat Zhou J, Zhang L, Wang M, Zhou L, Feng X, Yu L, et al. CPX targeting DJ-1 triggers ROS-induced cell death and protective autophagy in colorectal Cancer. Theranostics. 2019;9:5577–94.PubMedPubMedCentral Zhou J, Zhang L, Wang M, Zhou L, Feng X, Yu L, et al. CPX targeting DJ-1 triggers ROS-induced cell death and protective autophagy in colorectal Cancer. Theranostics. 2019;9:5577–94.PubMedPubMedCentral
7.
Zurück zum Zitat Lv H, Wang C, Fang T, Li T, Lv G, Han Q, et al. Vitamin C preferentially kills cancer stem cells in hepatocellular carcinoma via SVCT-2. NPJ Precis Oncol. 2018;2:1.PubMedPubMedCentral Lv H, Wang C, Fang T, Li T, Lv G, Han Q, et al. Vitamin C preferentially kills cancer stem cells in hepatocellular carcinoma via SVCT-2. NPJ Precis Oncol. 2018;2:1.PubMedPubMedCentral
8.
Zurück zum Zitat Shin B, Feser R, Nault B, Hunter S, Maiti S, Ugwuagbo KC, et al. miR526b and miR655 Induce Oxidative Stress in Breast Cancer. Int J Mol Sci. 2019;20. Shin B, Feser R, Nault B, Hunter S, Maiti S, Ugwuagbo KC, et al. miR526b and miR655 Induce Oxidative Stress in Breast Cancer. Int J Mol Sci. 2019;20.
9.
Zurück zum Zitat Cui J, Zhou Z, Yang H, Jiao F, Li N, Gao Y, et al. MST1 suppresses pancreatic Cancer progression via ROS-induced Pyroptosis. Mol Cancer Res. 2019;17:1316–25.PubMed Cui J, Zhou Z, Yang H, Jiao F, Li N, Gao Y, et al. MST1 suppresses pancreatic Cancer progression via ROS-induced Pyroptosis. Mol Cancer Res. 2019;17:1316–25.PubMed
10.
Zurück zum Zitat Guo H, Xu J, Zheng Q, He J, Zhou W, Wang K, et al. NRF2 SUMOylation promotes de novo serine synthesis and maintains HCC tumorigenesis. Cancer Lett. 2019;466:39–48.PubMed Guo H, Xu J, Zheng Q, He J, Zhou W, Wang K, et al. NRF2 SUMOylation promotes de novo serine synthesis and maintains HCC tumorigenesis. Cancer Lett. 2019;466:39–48.PubMed
11.
Zurück zum Zitat Yang WC, Tsai WC, Lin PM, Yang MY, Liu YC, Chang CS, et al. Human BDH2, an anti-apoptosis factor, is a novel poor prognostic factor for de novo cytogenetically normal acute myeloid leukemia. J Biomed Sci. 2013;20:58.PubMedPubMedCentral Yang WC, Tsai WC, Lin PM, Yang MY, Liu YC, Chang CS, et al. Human BDH2, an anti-apoptosis factor, is a novel poor prognostic factor for de novo cytogenetically normal acute myeloid leukemia. J Biomed Sci. 2013;20:58.PubMedPubMedCentral
12.
Zurück zum Zitat Guo K, Lukacik P, Papagrigoriou E, Meier M, Lee WH, Adamski J, et al. Characterization of human DHRS6, an orphan short chain dehydrogenase/reductase enzyme: a novel, cytosolic type 2 R-beta-hydroxybutyrate dehydrogenase. J Biol Chem. 2006;281:10291–7.PubMed Guo K, Lukacik P, Papagrigoriou E, Meier M, Lee WH, Adamski J, et al. Characterization of human DHRS6, an orphan short chain dehydrogenase/reductase enzyme: a novel, cytosolic type 2 R-beta-hydroxybutyrate dehydrogenase. J Biol Chem. 2006;281:10291–7.PubMed
13.
Zurück zum Zitat Devireddy LR, Hart DO, Goetz DH, Green MR. A mammalian siderophore synthesized by an enzyme with a bacterial homolog involved in enterobactin production. Cell. 2010;141:1006–17.PubMedPubMedCentral Devireddy LR, Hart DO, Goetz DH, Green MR. A mammalian siderophore synthesized by an enzyme with a bacterial homolog involved in enterobactin production. Cell. 2010;141:1006–17.PubMedPubMedCentral
14.
Zurück zum Zitat Zang W, Wang T, Wang Y, Chen X, Du Y, Sun Q, et al. Knockdown of long non-coding RNA TP73-AS1 inhibits cell proliferation and induces apoptosis in esophageal squamous cell carcinoma. Oncotarget. 2016;7:19960–74.PubMedPubMedCentral Zang W, Wang T, Wang Y, Chen X, Du Y, Sun Q, et al. Knockdown of long non-coding RNA TP73-AS1 inhibits cell proliferation and induces apoptosis in esophageal squamous cell carcinoma. Oncotarget. 2016;7:19960–74.PubMedPubMedCentral
15.
Zurück zum Zitat Liu JZ, Hu YL, Feng Y, Guo YB, Liu YF, Yang JL, et al. Rafoxanide promotes apoptosis and autophagy of gastric cancer cells by suppressing PI3K /Akt/mTOR pathway. Exp Cell Res. 2019;385:111691.PubMed Liu JZ, Hu YL, Feng Y, Guo YB, Liu YF, Yang JL, et al. Rafoxanide promotes apoptosis and autophagy of gastric cancer cells by suppressing PI3K /Akt/mTOR pathway. Exp Cell Res. 2019;385:111691.PubMed
16.
Zurück zum Zitat Hu YL, Feng Y, Ma P, Wang F, Huang H, Guo YB, et al. HAX-1 promotes the migration and invasion of hepatocellular carcinoma cells through the induction of epithelial-mesenchymal transition via the NF-kappaB pathway. Exp Cell Res. 2019;381:66–76.PubMed Hu YL, Feng Y, Ma P, Wang F, Huang H, Guo YB, et al. HAX-1 promotes the migration and invasion of hepatocellular carcinoma cells through the induction of epithelial-mesenchymal transition via the NF-kappaB pathway. Exp Cell Res. 2019;381:66–76.PubMed
17.
Zurück zum Zitat Zhao Q, Liu Y, Zhong J, Bi Y, Liu Y, Ren Z, et al. Pristimerin induces apoptosis and autophagy via activation of ROS/ASK1/JNK pathway in human breast cancer in vitro and in vivo. Cell Death Dis. 2019;5:125. Zhao Q, Liu Y, Zhong J, Bi Y, Liu Y, Ren Z, et al. Pristimerin induces apoptosis and autophagy via activation of ROS/ASK1/JNK pathway in human breast cancer in vitro and in vivo. Cell Death Dis. 2019;5:125.
18.
Zurück zum Zitat Kaminskyy VO, Piskunova T, Zborovskaya IB, Tchevkina EM, Zhivotovsky B. Suppression of basal autophagy reduces lung cancer cell proliferation and enhances caspase-dependent and -independent apoptosis by stimulating ROS formation. Autophagy. 2012;8:1032–44.PubMedPubMedCentral Kaminskyy VO, Piskunova T, Zborovskaya IB, Tchevkina EM, Zhivotovsky B. Suppression of basal autophagy reduces lung cancer cell proliferation and enhances caspase-dependent and -independent apoptosis by stimulating ROS formation. Autophagy. 2012;8:1032–44.PubMedPubMedCentral
19.
Zurück zum Zitat Fernandez-Gil BI, Guerra-Librero A, Shen YQ, Florido J, Martinez-Ruiz L, Garcia-Lopez S, et al. Melatonin enhances Cisplatin and radiation cytotoxicity in head and neck squamous cell carcinoma by stimulating mitochondrial ROS generation, apoptosis, and autophagy. Oxidative Med Cell Longev. 2019;2019:7187128. Fernandez-Gil BI, Guerra-Librero A, Shen YQ, Florido J, Martinez-Ruiz L, Garcia-Lopez S, et al. Melatonin enhances Cisplatin and radiation cytotoxicity in head and neck squamous cell carcinoma by stimulating mitochondrial ROS generation, apoptosis, and autophagy. Oxidative Med Cell Longev. 2019;2019:7187128.
20.
Zurück zum Zitat Kensler TW, Wakabayashi N, Biswal S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol. 2007;47:89–116.PubMed Kensler TW, Wakabayashi N, Biswal S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol. 2007;47:89–116.PubMed
21.
Zurück zum Zitat Jia J, Zhang HB, Shi Q, Yang C, Ma JB, Jin B, et al. KLF5 downregulation desensitizes castration-resistant prostate cancer cells to docetaxel by increasing BECN1 expression and inducing cell autophagy. Theranostics. 2019;9:5464–77.PubMedPubMedCentral Jia J, Zhang HB, Shi Q, Yang C, Ma JB, Jin B, et al. KLF5 downregulation desensitizes castration-resistant prostate cancer cells to docetaxel by increasing BECN1 expression and inducing cell autophagy. Theranostics. 2019;9:5464–77.PubMedPubMedCentral
22.
Zurück zum Zitat Wang XY, Zhang XH, Peng L, Liu Z, Yang YX, He ZX, et al. Bardoxolone methyl (CDDO-me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. Am J Transl Res. 2017;9:4652–72.PubMedPubMedCentral Wang XY, Zhang XH, Peng L, Liu Z, Yang YX, He ZX, et al. Bardoxolone methyl (CDDO-me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. Am J Transl Res. 2017;9:4652–72.PubMedPubMedCentral
23.
Zurück zum Zitat Baj J, Korona-Glowniak I, Forma A, Maani A, Sitarz E, Rahnama-Hezavah M, et al. Mechanisms of the epithelial-Mesenchymal transition and tumor microenvironment in helicobacter pylori-induced gastric Cancer. Cells. 2020;9. Baj J, Korona-Glowniak I, Forma A, Maani A, Sitarz E, Rahnama-Hezavah M, et al. Mechanisms of the epithelial-Mesenchymal transition and tumor microenvironment in helicobacter pylori-induced gastric Cancer. Cells. 2020;9.
24.
Zurück zum Zitat Yamaji Y, Hirata Y. Treatment for helicobacter pylori appears to reduce the incidence of gastric cancer: eradication effect or screening effect? Gut. 2020;69:605–6.PubMed Yamaji Y, Hirata Y. Treatment for helicobacter pylori appears to reduce the incidence of gastric cancer: eradication effect or screening effect? Gut. 2020;69:605–6.PubMed
25.
Zurück zum Zitat Zughaier SM, Stauffer BB, McCarty NA. Inflammation and ER stress downregulate BDH2 expression and dysregulate intracellular iron in macrophages. J Immunol Res. 2014;2014:140728.PubMedPubMedCentral Zughaier SM, Stauffer BB, McCarty NA. Inflammation and ER stress downregulate BDH2 expression and dysregulate intracellular iron in macrophages. J Immunol Res. 2014;2014:140728.PubMedPubMedCentral
26.
Zurück zum Zitat Chang LC, Chiang SK, Chen SE, Yu YL, Chou RH, Chang WC. Heme oxygenase-1 mediates BAY 11-7085 induced ferroptosis. Cancer Lett. 2018;416:124–37.PubMed Chang LC, Chiang SK, Chen SE, Yu YL, Chou RH, Chang WC. Heme oxygenase-1 mediates BAY 11-7085 induced ferroptosis. Cancer Lett. 2018;416:124–37.PubMed
27.
Zurück zum Zitat Liu Z, Velpula KK, Devireddy L. 3-Hydroxybutyrate dehydrogenase-2 and ferritin-H synergistically regulate intracellular iron. FEBS J. 2014;281:2410–21.PubMed Liu Z, Velpula KK, Devireddy L. 3-Hydroxybutyrate dehydrogenase-2 and ferritin-H synergistically regulate intracellular iron. FEBS J. 2014;281:2410–21.PubMed
28.
Zurück zum Zitat Jia J, Abudu YP, Claude-Taupin A, Gu Y, Kumar S, Choi SW, et al. Galectins control MTOR and AMPK in response to lysosomal damage to induce autophagy. Autophagy. 2019;15:169–71.PubMed Jia J, Abudu YP, Claude-Taupin A, Gu Y, Kumar S, Choi SW, et al. Galectins control MTOR and AMPK in response to lysosomal damage to induce autophagy. Autophagy. 2019;15:169–71.PubMed
29.
Zurück zum Zitat Shiloh R, Gilad Y, Ber Y, Eisenstein M, Aweida D, Bialik S, et al. Non-canonical activation of DAPK2 by AMPK constitutes a new pathway linking metabolic stress to autophagy. Nat Commun. 2018;9:1759.PubMedPubMedCentral Shiloh R, Gilad Y, Ber Y, Eisenstein M, Aweida D, Bialik S, et al. Non-canonical activation of DAPK2 by AMPK constitutes a new pathway linking metabolic stress to autophagy. Nat Commun. 2018;9:1759.PubMedPubMedCentral
30.
Zurück zum Zitat Sui X, Kong N, Ye L, Han W, Zhou J, Zhang Q, et al. p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett. 2014;344:174–9.PubMed Sui X, Kong N, Ye L, Han W, Zhou J, Zhang Q, et al. p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett. 2014;344:174–9.PubMed
31.
Zurück zum Zitat Liang H, Xiong Z, Li R, Hu K, Cao M, Yang J, et al. BDH2 is downregulated in hepatocellular carcinoma and acts as a tumor suppressor regulating cell apoptosis and autophagy. J Cancer. 2019;10:3735–45.PubMedPubMedCentral Liang H, Xiong Z, Li R, Hu K, Cao M, Yang J, et al. BDH2 is downregulated in hepatocellular carcinoma and acts as a tumor suppressor regulating cell apoptosis and autophagy. J Cancer. 2019;10:3735–45.PubMedPubMedCentral
32.
Zurück zum Zitat Fu R, Yang P, Li Z, Liu W, Amin S, Li Z. Avenanthramide a triggers potent ROS-mediated anti-tumor effects in colorectal cancer by directly targeting DDX3. Cell Death Dis. 2019;10:593.PubMedPubMedCentral Fu R, Yang P, Li Z, Liu W, Amin S, Li Z. Avenanthramide a triggers potent ROS-mediated anti-tumor effects in colorectal cancer by directly targeting DDX3. Cell Death Dis. 2019;10:593.PubMedPubMedCentral
33.
Zurück zum Zitat Sunilkumar D, Drishya G, Chandrasekharan A, Shaji SK, Bose C, Jossart J, et al. Oxyresveratrol drives caspase-independent apoptosis-like cell death in MDA-MB-231 breast cancer cells through the induction of ROS. Biochem Pharmacol. 2019;113724. Sunilkumar D, Drishya G, Chandrasekharan A, Shaji SK, Bose C, Jossart J, et al. Oxyresveratrol drives caspase-independent apoptosis-like cell death in MDA-MB-231 breast cancer cells through the induction of ROS. Biochem Pharmacol. 2019;113724.
34.
Zurück zum Zitat Holze C, Michaudel C, Mackowiak C, Haas DA, Benda C, Hubel P, et al. Oxeiptosis, a ROS-induced caspase-independent apoptosis-like cell-death pathway. Nat Immunol. 2018;19:130–40.PubMed Holze C, Michaudel C, Mackowiak C, Haas DA, Benda C, Hubel P, et al. Oxeiptosis, a ROS-induced caspase-independent apoptosis-like cell-death pathway. Nat Immunol. 2018;19:130–40.PubMed
35.
Zurück zum Zitat Zhang L, Wang H, Xu J, Zhu J, Ding K. Inhibition of cathepsin S induces autophagy and apoptosis in human glioblastoma cell lines through ROS-mediated PI3K/AKT/mTOR/p70S6K and JNK signaling pathways. Toxicol Lett. 2014;228:248–59.PubMed Zhang L, Wang H, Xu J, Zhu J, Ding K. Inhibition of cathepsin S induces autophagy and apoptosis in human glioblastoma cell lines through ROS-mediated PI3K/AKT/mTOR/p70S6K and JNK signaling pathways. Toxicol Lett. 2014;228:248–59.PubMed
36.
Zurück zum Zitat Yuan L, Wei S, Wang J, Liu X. Isoorientin induces apoptosis and autophagy simultaneously by reactive oxygen species (ROS)-related p53, PI3K/Akt, JNK, and p38 signaling pathways in HepG2 cancer cells. J Agric Food Chem. 2014;62:5390–400.PubMed Yuan L, Wei S, Wang J, Liu X. Isoorientin induces apoptosis and autophagy simultaneously by reactive oxygen species (ROS)-related p53, PI3K/Akt, JNK, and p38 signaling pathways in HepG2 cancer cells. J Agric Food Chem. 2014;62:5390–400.PubMed
37.
Zurück zum Zitat Ge W, Zhao K, Wang X, Li H, Yu M, He M, et al. iASPP is an Antioxidative factor and drives Cancer growth and drug resistance by competing with Nrf2 for Keap1 binding. Cancer Cell. 2017;32:561–73 e6.PubMed Ge W, Zhao K, Wang X, Li H, Yu M, He M, et al. iASPP is an Antioxidative factor and drives Cancer growth and drug resistance by competing with Nrf2 for Keap1 binding. Cancer Cell. 2017;32:561–73 e6.PubMed
38.
Zurück zum Zitat Liu K, Wang X, Sha K, Zhang F, Xiong F, Wang X, et al. Nuclear protein HMGN2 attenuates pyocyanin-induced oxidative stress via Nrf2 signaling and inhibits Pseudomonas aeruginosa internalization in A549 cells. Free Radic Biol Med. 2017;108:404–17.PubMed Liu K, Wang X, Sha K, Zhang F, Xiong F, Wang X, et al. Nuclear protein HMGN2 attenuates pyocyanin-induced oxidative stress via Nrf2 signaling and inhibits Pseudomonas aeruginosa internalization in A549 cells. Free Radic Biol Med. 2017;108:404–17.PubMed
39.
Zurück zum Zitat Eren E, Tufekci KU, Isci KB, Tastan B, Genc K, Genc S. Sulforaphane inhibits lipopolysaccharide-induced inflammation, cytotoxicity, oxidative stress, and miR-155 expression and switches to Mox phenotype through activating extracellular signal-regulated kinase 1/2-nuclear factor Erythroid 2-related factor 2/antioxidant response element pathway in murine microglial cells. Front Immunol. 2018;9:36.PubMedPubMedCentral Eren E, Tufekci KU, Isci KB, Tastan B, Genc K, Genc S. Sulforaphane inhibits lipopolysaccharide-induced inflammation, cytotoxicity, oxidative stress, and miR-155 expression and switches to Mox phenotype through activating extracellular signal-regulated kinase 1/2-nuclear factor Erythroid 2-related factor 2/antioxidant response element pathway in murine microglial cells. Front Immunol. 2018;9:36.PubMedPubMedCentral
40.
Zurück zum Zitat Kanlaya R, Khamchun S, Kapincharanon C, Thongboonkerd V. Protective effect of epigallocatechin-3-gallate (EGCG) via Nrf2 pathway against oxalate-induced epithelial mesenchymal transition (EMT) of renal tubular cells. Sci Rep. 2016;6:30233.PubMedPubMedCentral Kanlaya R, Khamchun S, Kapincharanon C, Thongboonkerd V. Protective effect of epigallocatechin-3-gallate (EGCG) via Nrf2 pathway against oxalate-induced epithelial mesenchymal transition (EMT) of renal tubular cells. Sci Rep. 2016;6:30233.PubMedPubMedCentral
41.
Zurück zum Zitat Kobayashi A, Kang MI, Okawa H, Ohtsuji M, Zenke Y, Chiba T, et al. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol Cell Biol. 2004;24:7130–9.PubMedPubMedCentral Kobayashi A, Kang MI, Okawa H, Ohtsuji M, Zenke Y, Chiba T, et al. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol Cell Biol. 2004;24:7130–9.PubMedPubMedCentral
42.
Zurück zum Zitat McMahon M, Itoh K, Yamamoto M, Hayes JD. Keap1-dependent proteasomal degradation of transcription factor Nrf2 contributes to the negative regulation of antioxidant response element-driven gene expression. J Biol Chem. 2003;278:21592–600.PubMed McMahon M, Itoh K, Yamamoto M, Hayes JD. Keap1-dependent proteasomal degradation of transcription factor Nrf2 contributes to the negative regulation of antioxidant response element-driven gene expression. J Biol Chem. 2003;278:21592–600.PubMed
43.
Zurück zum Zitat Nguyen T, Sherratt PJ, Huang HC, Yang CS, Pickett CB. Increased protein stability as a mechanism that enhances Nrf2-mediated transcriptional activation of the antioxidant response element. Degradation of Nrf2 by the 26 S proteasome. J Biol Chem. 2003;278:4536–41.PubMed Nguyen T, Sherratt PJ, Huang HC, Yang CS, Pickett CB. Increased protein stability as a mechanism that enhances Nrf2-mediated transcriptional activation of the antioxidant response element. Degradation of Nrf2 by the 26 S proteasome. J Biol Chem. 2003;278:4536–41.PubMed
44.
Zurück zum Zitat Shen H, Yang Y, Xia S, Rao B, Zhang J, Wang J. Blockage of Nrf2 suppresses the migration and invasion of esophageal squamous cell carcinoma cells in hypoxic microenvironment. Dis Esophagus. 2014;27:685–92.PubMed Shen H, Yang Y, Xia S, Rao B, Zhang J, Wang J. Blockage of Nrf2 suppresses the migration and invasion of esophageal squamous cell carcinoma cells in hypoxic microenvironment. Dis Esophagus. 2014;27:685–92.PubMed
45.
Zurück zum Zitat Gjyshi O, Bottero V, Veettil MV, Dutta S, Singh VV, Chikoti L, et al. Kaposi's sarcoma-associated herpesvirus induces Nrf2 during de novo infection of endothelial cells to create a microenvironment conducive to infection. PLoS Pathog. 2014;10:e1004460.PubMedPubMedCentral Gjyshi O, Bottero V, Veettil MV, Dutta S, Singh VV, Chikoti L, et al. Kaposi's sarcoma-associated herpesvirus induces Nrf2 during de novo infection of endothelial cells to create a microenvironment conducive to infection. PLoS Pathog. 2014;10:e1004460.PubMedPubMedCentral
46.
Zurück zum Zitat Yang X, Wang D, Ma Y, Xu X, Zhu Z, Wang X, et al. Continuous activation of Nrf2 and its target antioxidant enzymes leads to arsenite-induced malignant transformation of human bronchial epithelial cells. Toxicol Appl Pharmacol. 2015;289:231–9.PubMed Yang X, Wang D, Ma Y, Xu X, Zhu Z, Wang X, et al. Continuous activation of Nrf2 and its target antioxidant enzymes leads to arsenite-induced malignant transformation of human bronchial epithelial cells. Toxicol Appl Pharmacol. 2015;289:231–9.PubMed
47.
Zurück zum Zitat Lignitto L, LeBoeuf SE, Homer H, Jiang S, Askenazi M, Karakousi TR, et al. Nrf2 activation promotes lung Cancer metastasis by inhibiting the degradation of Bach1. Cell. 2019;178:316–29 e18.PubMed Lignitto L, LeBoeuf SE, Homer H, Jiang S, Askenazi M, Karakousi TR, et al. Nrf2 activation promotes lung Cancer metastasis by inhibiting the degradation of Bach1. Cell. 2019;178:316–29 e18.PubMed
48.
Zurück zum Zitat Nioi P, Nguyen T. A mutation of Keap1 found in breast cancer impairs its ability to repress Nrf2 activity. Biochem Biophys Res Commun. 2007;362:816–21.PubMed Nioi P, Nguyen T. A mutation of Keap1 found in breast cancer impairs its ability to repress Nrf2 activity. Biochem Biophys Res Commun. 2007;362:816–21.PubMed
49.
Zurück zum Zitat Perera RM, Bardeesy N. Cancer: when antioxidants are bad. Nature. 2011;475:43–4.PubMed Perera RM, Bardeesy N. Cancer: when antioxidants are bad. Nature. 2011;475:43–4.PubMed
50.
Zurück zum Zitat Homma S, Ishii Y, Morishima Y, Yamadori T, Matsuno Y, Haraguchi N, et al. Nrf2 enhances cell proliferation and resistance to anticancer drugs in human lung cancer. Clin Cancer Res. 2009;15:3423–32.PubMed Homma S, Ishii Y, Morishima Y, Yamadori T, Matsuno Y, Haraguchi N, et al. Nrf2 enhances cell proliferation and resistance to anticancer drugs in human lung cancer. Clin Cancer Res. 2009;15:3423–32.PubMed
51.
Zurück zum Zitat Hu XF, Yao J, Gao SG, Wang XS, Peng XQ, Yang YT, et al. Nrf2 overexpression predicts prognosis and 5-FU resistance in gastric cancer. Asian Pac J Cancer Prev. 2013;14:5231–5.PubMed Hu XF, Yao J, Gao SG, Wang XS, Peng XQ, Yang YT, et al. Nrf2 overexpression predicts prognosis and 5-FU resistance in gastric cancer. Asian Pac J Cancer Prev. 2013;14:5231–5.PubMed
52.
Zurück zum Zitat Li X, Liang M, Jiang J, He R, Wang M, Guo X, et al. Combined inhibition of autophagy and Nrf2 signaling augments bortezomib-induced apoptosis by increasing ROS production and ER stress in pancreatic cancer cells. Int J Biol Sci. 2018;14:1291–305.PubMedPubMedCentral Li X, Liang M, Jiang J, He R, Wang M, Guo X, et al. Combined inhibition of autophagy and Nrf2 signaling augments bortezomib-induced apoptosis by increasing ROS production and ER stress in pancreatic cancer cells. Int J Biol Sci. 2018;14:1291–305.PubMedPubMedCentral
53.
Zurück zum Zitat Lim J, Lee SH, Cho S, Lee IS, Kang BY, Choi HJ. 4-methoxychalcone enhances cisplatin-induced oxidative stress and cytotoxicity by inhibiting the Nrf2/ARE-mediated defense mechanism in A549 lung cancer cells. Mol Cell. 2013;36:340–6. Lim J, Lee SH, Cho S, Lee IS, Kang BY, Choi HJ. 4-methoxychalcone enhances cisplatin-induced oxidative stress and cytotoxicity by inhibiting the Nrf2/ARE-mediated defense mechanism in A549 lung cancer cells. Mol Cell. 2013;36:340–6.
Metadaten
Titel
BDH2 triggers ROS-induced cell death and autophagy by promoting Nrf2 ubiquitination in gastric cancer
verfasst von
Jia-Zhou Liu
Yi-Lin Hu
Ying Feng
Yun Jiang
Yi-Bing Guo
Yi-Fei Liu
Xi Chen
Jun-Ling Yang
Yu-yan Chen
Qin-Sheng Mao
Wan-Jiang Xue
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2020
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-020-01620-z

Weitere Artikel der Ausgabe 1/2020

Journal of Experimental & Clinical Cancer Research 1/2020 Zur Ausgabe

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.