Introduction
Prostate cancer (PCa)
4The abbreviations used are:
PCa
prostate cancer
BMA1
bafilomycin A1
CDDP
cisplatin
DOX
doxorubicin
Dub
deubiquitinase
EMT
epithelial to mesenchymal transition
IHC
immunohistochemistry
MTT
3-(4,5-dimethylthylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
PBX1
pre-B cell leukemia homeobox-1
UPP
ubiquitin-proteasome pathway
USP9x
ubiquitin-specific protease 9, X-linked
PARP
poly(ADP-ribose) polymerase
CHX
cycloheximide
PI
propidium iodide
HA
hemagglutinin
IP
immunoprecipitation
IB
immunoblot
GAPDH
glyceraldehyde-3-phosphate dehydrogenase
Ub
ubiquitin
BPH
benign prostate hyperplasia
PIN
prostatic intraepithelial neoplasia.
is a malignant disease developed in the prostate, a gland in the male reproductive system. The epidemiological studies reveal that PCa is one of the most common cancers in men and one of the leading causes of cancer-related deaths worldwide (
1- Chang A.J.
- Autio K.A.
- Roach 3rd, M.
- Scher H.I.
High-risk prostate cancer-classification and therapy.
). Several treatment modalities have been developed against prostate cancers, including androgen-deprivation therapy, localized radiotherapy and chemotherapy. Currently, androgen-deprivation therapy is the standard frontline therapy for advanced PCa patients; nevertheless, most patients will eventually develop resistance and these castration-resistant PCa patients rely on chemotherapy. Unfortunately, this regimen only prolongs modest survival of PCa patients, of which many acquire chemoresistance and eventually evolve to a fatal clinical outcome (
1- Chang A.J.
- Autio K.A.
- Roach 3rd, M.
- Scher H.I.
High-risk prostate cancer-classification and therapy.
). It is widely believed that molecular and genetic events are key players in the resistance but they are not well defined.
Pre-B cell leukemia homeobox-1 (PBX1), a member of the TALE (three-amino acid loop extension) family of atypical homeodomain proteins, is cloned from pre-B cell leukemia (
2- Kamps M.P.
- Look A.T.
- Baltimore D.
The human t(1;19) translocation in pre-B ALL produces multiple nuclear E2A-Pbx1 fusion proteins with differing transforming potentials.
), our recent study demonstrated that it up-regulates the transcription of ring finger protein 6 and contributes to leukemia chemoresistance (
3- Xu X.
- Han K.
- Tang X.
- Zeng Y.
- Lin X.
- Zhao Y.
- Zhang Z.
- Cao B.
- Wu D.
- Mao X.
The ring finger protein RNF6 induces leukemia cell proliferation as a direct target of pre-B-cell leukemia homeobox 1.
). However, more and more evidence shows that PBX1 is dysregulated and contributes to proliferation, survival, metastasis, and chemoresistance in various solid tumors, including breast, lung, gastric, and ovarian cancers. For example, high expression of PBX1 drives breast cancer proliferation and metastasis by regulating the estrogen receptor transcriptional response to epidermal growth factor signaling (
4- Magnani L.
- Patten D.K.
- Nguyen V.T.
- Hong S.P.
- Steel J.H.
- Patel N.
- Lombardo Y.
- Faronato M.
- Gomes A.R.
- Woodley L.
- Page K.
- Guttery D.
- Primrose L.
- Fernandez Garcia D.
- et al.
The pioneer factor PBX1 is a novel driver of metastatic progression in ERα-positive breast cancer.
). In gastric carcinomas (
5- He C.
- Wang Z.
- Zhang L.
- Yang L.
- Li J.
- Chen X.
- Zhang J.
- Chang Q.
- Yu Y.
- Liu B.
- Zhu Z.
A hydrophobic residue in the TALE homeodomain of PBX1 promotes epithelial-to-mesenchymal transition of gastric carcinoma.
) and nonsmall cell lung cancers (
6- Risolino M.
- Mandia N.
- Iavarone F.
- Dardaei L.
- Longobardi E.
- Fernandez S.
- Talotta F.
- Bianchi F.
- Pisati F.
- Spaggiari L.
- Harter P.N.
- Mittelbronn M.
- Schulte D.
- Incoronato M.
- Di Fiore P.P.
- Blasi F.
- Verde P.
Transcription factor PREP1 induces EMT and metastasis by controlling the TGF-β-SMAD3 pathway in non-small cell lung adenocarcinoma.
), PBX1 promotes epithelial to mesenchymal transition that is associated with chemoresistance. Although there are less studies on PBX1 in PCa, a recent study revealed that PBX3, a homolog of PBX1, is a putative biomarker of aggressive PCa (
7- Ramberg H.
- Grytli H.H.
- Nygård S.
- Wang W.
- Ögren O.
- Zhao S.
- Løvf M.
- Katz B.
- Skotheim R.I.
- Bjartell A.
- Eri L.M.
- Berge V.
- Svindland A.
- Taskén K.A.
PBX3 is a putative biomarker of aggressive prostate cancer.
). There are two PBX1 transcripts: PBX1a and PBX1b, between which the only difference is that PBX1b lacks the 7th exon (
8- Park J.T.
- Shih IeM.
- Wang T.L.
Identification of Pbx1, a potential oncogene, as a Notch3 target gene in ovarian cancer.
), but their biological functions remain very similar as a transcription factor.
Protein stability is modulated by two major metabolizing pathways in eukaryotes, one is the autophagy-lysosomal pathway, and the other is the ubiquitin-proteasomal pathway (UPP). The turnover of most transcription factors is processed via UPP (
9- Sacco J.J.
- Coulson J.M.
- Clague M.J.
- Urbé S.
Emerging roles of deubiquitinases in cancer-associated pathways.
). Proteasomal degradation depends on protein ubiquitination, a process in which a ubiquitin chain is attached to a specific lysine residue at the substrate protein to be degraded. Notably, protein ubiquitination is a dynamic process and the covalent ubiquitin molecules could be removed by certain deubiquitinases (Dubs). Ubiquitin-specific protease 9, X-linked (USP9x), one of the most studies Dubs, has been found to prevent ubiquitination from diverse protein substrates (
10- Théard D.
- Labarrade F.
- Partisani M.
- Milanini J.
- Sakagami H.
- Fon E.A.
- Wood S.A.
- Franco M.
- Luton F.
USP9x-mediated deubiquitination of EFA6 regulates de novo tight junction assembly.
,
11- Trivigno D.
- Essmann F.
- Huber S.M.
- Rudner J.
Deubiquitinase USP9x confers radioresistance through stabilization of Mcl-1.
). In the present study, we found that USP9x stabilizes PBX1 by preventing its polyubiquitination and confers to PCa proliferation. Moreover, we found that inhibition of the USP9x/PBX1 axis overcomes PCa chemoresistance.
Discussion
The above study demonstrated that as an oncogenic transcription factor PBX1 is highly expressed in most PCa tissues, promotes PCa cell proliferation, and confers to chemoresistance against representative anti-cancer drugs. Moreover, we found for the first time that USP9x is a deubiquitinase in stabilizing PBX1 by inhibiting its polyubiquitination. We also demonstrated that targeting the USP9x/PBX1 axis could be a potential modality to overcome PCa chemoresistance.
Resistance is a major obstacle to success with chemotherapy in advanced PCa, but the underlying mechanisms are not well known. Evidence has shown that chemoresistance of advanced PCa might be associated with the presence of cancer stem cells that regenerate tumorigenicity, overactivated androgen receptor signaling that promotes PCa cell proliferation, ABC proteins that pump out drugs outside the cancer cells, epithelial-mesenchymal transition (EMT) that confers to cancer cell migratory, and invasive ability (
17Profiling prostate cancer therapeutic resistance.
,
18- Zhang W.
- Meng Y.
- Liu N.
- Wen X.F.
- Yang T.
Insights into chemoresistance of prostate cancer.
). However, PCa cells are highly heterogeneous (
1- Chang A.J.
- Autio K.A.
- Roach 3rd, M.
- Scher H.I.
High-risk prostate cancer-classification and therapy.
), even in prostate tumors with similar pathology (
19- Boutros P.C.
- Fraser M.
- Harding N.J.
- de Borja R.
- Trudel D.
- Lalonde E.
- Meng A.
- Hennings-Yeomans P.H.
- McPherson A.
- Sabelnykova V.Y.
- Zia A.
- Fox N.S.
- Livingstone J.
- Shiah Y.J.
- Wang J.
- et al.
Spatial genomic heterogeneity within localized, multifocal prostate cancer.
). Therefore, in specific subsets of chemoresistant PCa patients, unique expression of certain genes might play a key role. In the present study, we found that PBX1 expression in prostate tissues is increased following the malignancy from BPH and PIN to PCa. Although we could not provide the PBX1 expression data from normal prostate tissues, PBX1 was found to be positive in one-third of BPH patients, which rose to around 54% in PIN patients, and it was found in more than 70% of PCa patients, suggesting that the PBX1 expression level might be involved in the progress of PCa. We also found PBX1 is highly expressed in three-fourths of PCa cell lines. Moreover, PBX1 expression determines PCa cell proliferation and drug sensitivity. Ectopic expression of PBX1 promotes PCa cell proliferation and confers to resistance to cytotoxic anti-cancer drugs, in contrast, knockdown of PBX1 restores drug sensitivity. Although it is originally identified from pre-B leukemia cells (
2- Kamps M.P.
- Look A.T.
- Baltimore D.
The human t(1;19) translocation in pre-B ALL produces multiple nuclear E2A-Pbx1 fusion proteins with differing transforming potentials.
), PBX1 is now also reported in other cancers such as lung, breast, and ovarian cancers to promote cancer cell proliferation, survival, and metastasis. PBX1 acts as a stem cell reprogramming factor in regulating long-term hematopoietic stem cells by maintaining their self-renewal and quiescence (
20- Ficara F.
- Murphy M.J.
- Lin M.
- Cleary M.L.
Pbx1 regulates self-renewal of long-term hematopoietic stem cells by maintaining their quiescence.
) but also driving stemness of ovarian cancer stem cells (
12- Jung J.G.
- Shih I.M.
- Park J.T.
- Gerry E.
- Kim T.H.
- Ayhan A.
- Handschuh K.
- Davidson B.
- Fader A.N.
- Selleri L.
- Wang T.L.
Ovarian cancer chemoresistance relies on the stem cell reprogramming factor PBX1.
) and initiating leukemogenesis (
21E2a/Pbx1 induces the rapid proliferation of stem cell factor-dependent murine pro-T cells that cause acute T-lymphoid or myeloid leukemias in mice.
). It is also believed to be a pioneer factor promoting a transcriptional program favorable to breast cancer progression (
22- Magnani L.
- Ballantyne E.B.
- Zhang X.
- Lupien M.
PBX1 genomic pioneer function drives ERα signaling underlying progression in breast cancer.
). Furthermore, PBX1 has been extensively involved in promoting EMT in various cancers in collaboration with other transcription factors such as ZEB2 (
23- Zhu X.
- Wei L.
- Bai Y.
- Wu S.
- Han S.
FoxC1 promotes epithelial-mesenchymal transition through PBX1 dependent transactivation of ZEB2 in esophageal cancer.
) and PREP1 (
6- Risolino M.
- Mandia N.
- Iavarone F.
- Dardaei L.
- Longobardi E.
- Fernandez S.
- Talotta F.
- Bianchi F.
- Pisati F.
- Spaggiari L.
- Harter P.N.
- Mittelbronn M.
- Schulte D.
- Incoronato M.
- Di Fiore P.P.
- Blasi F.
- Verde P.
Transcription factor PREP1 induces EMT and metastasis by controlling the TGF-β-SMAD3 pathway in non-small cell lung adenocarcinoma.
). Therefore, there is no doubt that PBX1 promotes PCa cell proliferation, survival, and chemoresistance. Actually, a recent study showed that ovarian cancer chemoresistance relies on PBX1 overexpression (
12- Jung J.G.
- Shih I.M.
- Park J.T.
- Gerry E.
- Kim T.H.
- Ayhan A.
- Handschuh K.
- Davidson B.
- Fader A.N.
- Selleri L.
- Wang T.L.
Ovarian cancer chemoresistance relies on the stem cell reprogramming factor PBX1.
). Our recent study has demonstrated that PBX1 promotes the expression of oncogenic RNF6, thus increasing leukemia chemoresistance to doxorubicin (
3- Xu X.
- Han K.
- Tang X.
- Zeng Y.
- Lin X.
- Zhao Y.
- Zhang Z.
- Cao B.
- Wu D.
- Mao X.
The ring finger protein RNF6 induces leukemia cell proliferation as a direct target of pre-B-cell leukemia homeobox 1.
). Based on previous reports and our present study, PBX1 is believed to confer to chemoresistance in PCa cells.
In the present study, we also found that PBX1 stability is modulated by the ubiquitin proteasomal system because the PBX1 protein could be polyubiquitinated and its degradation could be prevented by inhibition of proteasomes but not lysosomes. Moreover, the Dub USP9x interacts with PBX1 and abolishes its polyubiquitination level by reducing its K48-linked ubiquitin chain thus leading to PBX1 stabilization. Previous studies showed that USP9x ablates K48-linked ubiquitination of ERG, another oncogenic transcription factor (
24- Wang S.
- Kollipara R.K.
- Srivastava N.
- Li R.
- Ravindranathan P.
- Hernandez E.
- Freeman E.
- Humphries C.G.
- Kapur P.
- Lotan Y.
- Fazli L.
- Gleave M.E.
- Plymate S.R.
- Raj G.V.
- Hsieh J.T.
- Kittler R.
Ablation of the oncogenic transcription factor ERG by deubiquitinase inhibition in prostate cancer.
). In the present study, we also found that USP9x abolishes K63-linked ubiquitination from PBX1. The K48- and K63-linked ubiquitination forms have been reported in the same proteins, such as PTEN and ErbB2 (
25- Marx C.
- Held J.M.
- Gibson B.W.
- Benz C.C.
ErbB2 trafficking and degradation associated with K48 and K63 polyubiquitination.
), but the function may vary dependent on the specific ubiquitination types. It is well known that K48-linked polyubiquitination results in substrate degradation, whereas the K63-linked modification usually alters the function of the target proteins. In BCR-ABL expressing chronic myeloid leukemia cells, the USP9x inhibitor WP1130 leads to increased K63-linked ubiquitination of BCR-ABL that results in BCR-ABL accumulation in aggresomes in which it fails to conduct signal transduction, therefore chronic myeloid leukemia cells undergo apoptosis (
16- Sun H.
- Kapuria V.
- Peterson L.F.
- Fang D.
- Bornmann W.G.
- Bartholomeusz G.
- Talpaz M.
- Donato N.J.
Bcr-Abl ubiquitination and Usp9x inhibition block kinase signaling and promote CML cell apoptosis.
). In PCa cells, we found USP9x down-regulates both K48- and K63-linked ubiquitination levels of PBX1, suggesting these two modifications might enhance PBX1 oncogenicity, whereas inhibition of USP9x suppresses its activity. As shown in
Fig. 6, WP1130 leads to PBX1 degradation and reverses PCa cell chemoresistance. Notably, we also found that WP1130 leads to DU145 cell apoptosis although this cell line does not express PBX1. The possible reason could be that USP9x has diverse oncogenic substrate proteins and these proteins also promote PCa cell survival and proliferation. Actually, oncoproteins such as MCL-1 (
26- Schwickart M.
- Huang X.
- Lill J.R.
- Liu J.
- Ferrando R.
- French D.M.
- Maecker H.
- O'Rourke K.
- Bazan F.
- Eastham-Anderson J.
- Yue P.
- Dornan D.
- Huang D.C.
- Dixit V.M.
Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival.
) and YAP1 (
27- Li L.
- Liu T.
- Li Y.
- Wu C.
- Luo K.
- Yin Y.
- Chen Y.
- Nowsheen S.
- Wu J.
- Lou Z.
- Yuan J.
The deubiquitinase USP9X promotes tumor cell survival and confers chemoresistance through YAP1 stabilization.
) are also substrates of USP9x, and these two oncoproteins are also highly expressed in PCa and promote PCa cell proliferation and survival (
28- Lin C.L.
- Chen C.M.
- Lin C.L.
- Cheng C.W.
- Lee C.H.
- Hsieh Y.H.
Norcantharidin induces mitochondrial-dependent apoptosis through Mcl-1 inhibition in human prostate cancer cells.
,
29- Jiang N.
- Ke B.
- Hjort-Jensen K.
- Iglesias-Gato D.
- Wang Z.
- Chang P.
- Zhao Y.
- Niu X.
- Wu T.
- Peng B.
- Jiang M.
- Li X.
- Shang Z.
- Wang Q.
- Chang C.
- Flores-Morales A.
- Niu Y.
YAP1 regulates prostate cancer stem cell-like characteristics to promote castration resistant growth.
). These findings further demonstrated that USP9x plays a critical role in promoting PCa cell proliferation, survival, and chemoresistance. Inhibition of USP9x could directly lead to apoptosis of PCa cells that does not express PBX1, however, in PBX1-expressing PCa cells, inhibition of USP9x could help overcome their chemoresistance because WP1130 sensitizes PC3 cells to cisplatin at the same concentrations as shown in
Fig. 6.
In conclusion, the present study for the first time highlights the role of PBX1 in PCa chemoresistance and identifies USP9x as a Dub of PBX1. By stabilizing PBX1, USP9x also promotes PCa cell proliferation. Moreover, inhibition of USP9x sensitizes PBX1-expressing PCa cell apoptosis. Therefore, the present study suggests that targeting at the USP9x/PBX1 axis could be a potential modality to overcome PCa chemoresistance due to the high expression of PBX1.
Experimental procedures
Prostate tissues
Prostate tissues were obtained from patients with BPH, PIN, or PCa when patients made their first visits to the Department of Urology, Jinling Hospital, Nanjing Jinling Hospital, from May 2016 to April 2018. The usage of these specimens for the present study was informed to each patient and a signed content was obtained from each patient. The collection and use of prostate tissues in the present study was approved by the Review Board of Medical Ethics of Jinling Hospital in accordance with the Declaration of Helsinki.
Cell culture and chemicals
Human PCa cell lines (PC3 and DU145) were purchased from American Type Culture Collection (Manassas, VA). Cell lines 22RV1 and PC-3M were obtained from China Center for Type Culture Collection (Wuhan, China); human embryonic kidney (HEK293T) cell line was provided by Dr. Michael Moran, The University of Toronto, Canada. PCa cells were cultured in RPMI 1640 medium. HEK293T cells were maintained in Dulbecco’s high glucose modified Eagle’s medium. All the media were supplemented with 10% fetal bovine serum, 100 μg/ml of penicillin, and 100 units/ml of streptomycin.
Doxorubicin was purchased from Sangon Biotech Co., Ltd. (Shanghai, Shanghai, China). Cisplatin was purchased from Jiangsu Hansoh Pharmaceutical Group Co., Ltd. (Lianyungang, Jiangsu, China). WP1130 was purchased from Selleck Chemicals (Houston, TX). MG132 and bortezomib were purchased from Chemcatch (Shanghai, Shanghai, China) and Adamas Chemicals (Shanghai, Shanghai, China), respectively.
Antibodies and plasmids
Rabbit anti-PBX1, anti-caspase-3, anti-cleaved caspase-3, and anti-PARP antibodies were obtained from Cell Signaling Technology, Inc., (Danvers, MA). Anti-HA, anti-FLAG, and anti-Myc monoclonal antibodies were obtained from Medical & Biological Laboratories Co., Ltd. (Tokyo, Japan). A mouse anti-GAPDH antibody was purchased from Abgent (Suzhou, Jiangsu, China). A mouse anti-V5 antibody was purchased from Huabio Antibodies (Hangzhou, Zhejiang, China). The rabbit anti-USP9x antibody was obtained from Proteintech Group Co. (Wuhan, Hubei, China).
The PBX1a and ubiquitin (Ub) plasmids were prepared as described previously (
3- Xu X.
- Han K.
- Tang X.
- Zeng Y.
- Lin X.
- Zhao Y.
- Zhang Z.
- Cao B.
- Wu D.
- Mao X.
The ring finger protein RNF6 induces leukemia cell proliferation as a direct target of pre-B-cell leukemia homeobox 1.
) and subcloned into a pcDNA3.1 vector carrying HA, FLAG, or Myc tags. The V5-USP9x plasmid was generously provided by Dr. Guoqiang Xu (Soochow University). The PBX1b plasmid was purchased from General Biosystems, Inc. (Morrisville, NC).
Immunohistochemistry (IHC)
All tissues were embedded by paraffin waxes and were sliced to 5-μm thick sections with a rotary microtome (Leica Biosystems, Wetzlar, Germany) and mounted onto slides. Immunostaining was conducted using a Vectastain ABC kit (Vector Laboratories, Burlingame, CA) following the manufacturer’s instructions. Briefly, slides were deparaffinized, rehydrated, and treated with citric acid solution before being applied for IHC. After blocking the endogenous peroxidase activity by preincubation in 3% hydrogen peroxide solution, the slides were incubated in blocking solution (PBS, 3% BSA) and sequentially incubated with anti-PBX1 antibody (Abcam). The sections were counterstained with hematoxylin (Wuhan Service Biotechnology Co., Ltd., Wuhan, China) for nuclear staining. To evaluate the expression levels of the protein, the intensity of staining was scored according to a semiquantitative 4-grade scale as follows: 0, none (no positive tumor cells); 1, weak (<33% positive tumor cells); 2, moderate (33–67% positive tumor cells); 3, strong (>67% positive tumor cells). An intensity score of ≥2 was considered as high expression, whereas <2 was regarded as low expression.
Immunoblotting (IB)
Cell lysates were prepared as described previously (
30- Xu Y.
- Zhang Z.
- Li J.
- Tong J.
- Cao B.
- Taylor P.
- Tang X.
- Wu D.
- Moran M.F.
- Zeng Y.
- Mao X.
The ubiquitin-conjugating enzyme UBE2O modulates c-Maf stability and induces myeloma cell apoptosis.
). After concentration determination, equal amounts (40 μg) of total proteins were resolved by SDS-PAGE, followed by IB analyses with specific antibodies.
Immunoprecipitation (IP)
Twenty-four h after the cells were transfected with appropriate plasmids, cells were treated with MG132 for another 12 h. Cell lysates were incubated with the indicated antibodies overnight at 4 °C, followed by incubation with protein A/G–Sepharose beads (Beyotime Institute of Biotechnology, Nantong, Jiangsu, China) for 4 h. The proteins were then subjected to the IB assay with appropriate antibodies.
Cycloheximide (CHX) chase assay
HEK293T cells were transfected with HA-PBX1 and V5-USP9x plasmids for 24 h before being treated with CHX (100 μg/ml) for 0 to 12 h. Whole cell lysates were prepared for the IB assay with specific antibodies.
Luciferase assay
The RNF6 promoter containing a PBX1 recognition element (PBX1.Luci) was cloned into a pGL4 luciferase report system (Promega, Madison, WI) as described previously (
3- Xu X.
- Han K.
- Tang X.
- Zeng Y.
- Lin X.
- Zhao Y.
- Zhang Z.
- Cao B.
- Wu D.
- Mao X.
The ring finger protein RNF6 induces leukemia cell proliferation as a direct target of pre-B-cell leukemia homeobox 1.
). To examine the effect of USP9x on PBX1 transcriptional activity, PBX1.Luci and HA-PBX1a were co-transfected into HEK293T cells. Thirty-six h later, luciferase activity was analyzed with the Bright-Glo substrate (Promega) as described previously (
3- Xu X.
- Han K.
- Tang X.
- Zeng Y.
- Lin X.
- Zhao Y.
- Zhang Z.
- Cao B.
- Wu D.
- Mao X.
The ring finger protein RNF6 induces leukemia cell proliferation as a direct target of pre-B-cell leukemia homeobox 1.
).
Plasmid transfection
Prostate cancer lines were transfected with individual expressing plasmids including pcDNA3.1-PBX1a, -PBX1b, or -USP9x by Lipofectamine 2000® (ThermoFisher Scientific, Waltham, MA) according to the manufacturer’s instructions. HEK293T cells were transfected with specific plasmids as needed using polyethyleneimine (Sigma) as described previously (
30- Xu Y.
- Zhang Z.
- Li J.
- Tong J.
- Cao B.
- Taylor P.
- Tang X.
- Wu D.
- Moran M.F.
- Zeng Y.
- Mao X.
The ubiquitin-conjugating enzyme UBE2O modulates c-Maf stability and induces myeloma cell apoptosis.
). The appropriate amounts of plasmids were determined to result in a transfection efficiency about 70–80% when cells were split for further studies.
siRNA transfection
Small interfering RNAs (siRNAs) were transfected into PC3 or DU145 cells by Lipofectamine® 2000 as shown above. The specific siRNA sequences targeting PBX1 were as follows: siPBX1#1, 5′-CCA TCA CAG ACC AGA GTT T-3′; si-PBX1#2, 5′-GAA TGA AGC CTG CCT TGTT-3′; si-PBX1#3, 5′-GGA AGA GAC GGA ATT TCAA-3′. The specific sequence targeting USP9x was si-USP9x, 5′-ACA CGA UGC UUU AGA AUU UTT-3′. All the siRNAs were purchased from RiboBio Inc. (Guangzhou, Guangdong, China).
Cell viability
Cell viability was measured by the MTT (3-(4,5-dimethylthylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay as described previously (
31- Mao X.
- Hou T.
- Cao B.
- Wang W.
- Li Z.
- Chen S.
- Fei M.
- Hurren R.
- Gronda M.
- Wu D.
- Trudel S.
- Schimmer A.D.
The tricyclic antidepressant amitriptyline inhibits d-cyclin transactivation and induces myeloma cell apoptosis by inhibiting histone deacetylases: in vitro and in silico evidence.
) after transfection with specific plasmids or drug treatment.
Flow cytometric analysis
After treatment with appropriate drugs with or without plasmid transfections, PCa cells were stained with Annexin V and PI staining (MultiSciences Biotech Co. Ltd., Hangzhou, Zhejiang, China) for 5 min in the dark, followed by analyses on a flow cytometer (FACS Calibur®; BD Biosciences) as described previously (
31- Mao X.
- Hou T.
- Cao B.
- Wang W.
- Li Z.
- Chen S.
- Fei M.
- Hurren R.
- Gronda M.
- Wu D.
- Trudel S.
- Schimmer A.D.
The tricyclic antidepressant amitriptyline inhibits d-cyclin transactivation and induces myeloma cell apoptosis by inhibiting histone deacetylases: in vitro and in silico evidence.
).
Densitomestric analyses
Densitometric analyses of Western blots in the protein stability were performed as described previously (
3- Xu X.
- Han K.
- Tang X.
- Zeng Y.
- Lin X.
- Zhao Y.
- Zhang Z.
- Cao B.
- Wu D.
- Mao X.
The ring finger protein RNF6 induces leukemia cell proliferation as a direct target of pre-B-cell leukemia homeobox 1.
) using ImageJ® software developed by the National Institutes of Health.
Statistics
All experiments were performed at least 3 times except for the IHC studies. Statistical analyses between the control and the experimental groups were analyzed by the Student’s t test and the p value <0.05 was considered statistically significant.
Author contributions
Y. L. and X. X. data curation; Y. L. and G. S. formal analysis; Y. L., X. X., P. L., Y. H., Y. Z., B. C., and Z. Z. investigation; Y. L., X. X., Y. Z., B. C., Z. Z., and J. L. methodology; Y. L., G. S., J. L., and X. M. writing-review and editing; Z. Z. validation; J. L., X. Z., and X. M. project administration; X. Z. and X. M. funding acquisition; X. M. conceptualization; X. M. supervision; X. M. writing-original draft.
Article info
Publication history
Published online: February 04, 2019
Received in revised form:
January 29,
2019
Received:
September 29,
2018
Edited by Xiao-Fan Wang
Footnotes
This work was supported by National Natural Science Foundation of China Grants 81770154 (to X. M.), 30901716 (to X. X.), 81320108023 (to X. M.), and 81770215 (to B. C.), Natural Science Foundation of Jiangsu Higher Education Institutes of China Grant 17KJA180010 (to X. M.), the Priority Academic Program Development of Jiangsu Higher Education Institutions (to X. M.), and Jiangsu Key Laboratory of Neuropsychiatric Diseases Grant BK2013003 (to X. M.). The authors declare that they have no conflicts of interest with the contents of this article.
Copyright
© 2019 Liu et al.