Download as pdf or txt
Download as pdf or txt
You are on page 1of 42

Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.

CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Exosome-Transmitted PSMA3 and PSMA3-AS1 Promotes

Proteasome Inhibitors Resistance in Multiple Myeloma


Hongxia Xu1,2, +, Huiying Han1, +, Sha Song1, Nengjun Yi3, Chen’ao Qian4, Yingchun
Qiu1, Wenqi Zhou1, Yating Hong5, Wenyue Zhuang6, Zhengyi Li7, Bingzong Li5,*,
Wenzhuo Zhuang1, *
1
Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow
University, Suzhou, China.
2
Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang,
China.
3
Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL
35294, USA.
4
Department of Bioinformatics, School of Biology & Basic Medical Sciences,
Soochow University, Suzhou, China.
5
Department of Haematology, the Second Affiliated Hospital of Soochow University,
Suzhou, China.
6
Department of Molecular Biology, School of Laboratory Medicine of Beihua
University, Jilin, China.
7
Department of Clinical Examination Basis, Laboratory Acadamy of Jilin Medical
College, Jilin, China.

+
: Contribute equally to this work
*: Correspondence: Wenzhuo Zhuang, Email: zhuangwenzhuo@suda.edu.cn, Tel.:
86−512−65880103, Address: School of Biology & Basic Medical Sciences, Soochow
University, Ren Ai Road 199, Suzhou, 215123, China. Or Bingzong Li, Email:
lbzwz0907@hotmail.com, Tel.: 86−512−67784069, Address: The Second Affiliated
Hospital of Soochow University, San Xiang Road 1055, Suzhou, 215006, China.

Running title: Exosomal PSMA3 and PSMA3-AS1 in PIs resistance


Key words: Multiple myeloma, Proteasome inhibitors, Exosome, Long non-coding
RNA, Drug resistance

Financial support: This study was supported by Natural Science Foundation of


Jiangsu Province China (BK20161218, BK20161223), the Science and Technology
Development Project of Suzhou City (SS201856), National Natural Science
Foundation of China (81670191, 81673448), The Applied Basic Research Programs
of Suzhou City (SYS201546), Natural Science Foundation of Jilin Province
(20160101234JC).

Conflict of Interest statement: The authors declare no potential conflicts of interest.


Text word count: 4830, Number of figures: 6, Number of tables: 0.
1

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Translational Relevance

PIs resistance is a major challenge for MM. The bone marrow microenvironment

promotes myeloma cells survival and proliferation through the interaction between the

MSCs and MM cells. The bone marrow microenvironment promotes the interactions

between the MSCs and MM cells which permit MM to survive and proliferate. Here

we discovered that exosome-mediated transfer of PSMA3 (encodes proteasome

subunit α7) and lncPSMA3-AS1 from MSCs to MM cells contributed to PIs

resistance. PSMA3-AS1, which arises from the antisense strand of PSMA3, was

highly expressed in myeloma cells (r-MM) and MSCs (r-MSCs)-derived from

bortezomib-resistant patients. As a pair of protein-coding/non-coding antisense

transcripts, PSMA3 and PSMA3-AS1 were disordered concurrently and correlated

positively in MM cells, driving PIs sensitivity in MM. These results provided in vitro

and in vivo evidence that interference with exosome RNAs served as a promising

approach to overcome PIs resistance in MM. Moreover, our data indicated that

circulating exosomal PSMA3 and PSMA3-AS1 could develop the prognostic

stratification of MM patients, in addition to the international staging system.

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract
Purpose: How exosomal RNAs released within the bone marrow microenvironment

affect proteasome inhibitors (PIs) sensitivity of multiple myeloma (MM) is currently

unknown. This study aims to evaluate which exosomal RNAs are involved and by

which molecular mechanisms they exert this function.

Experimental Design: Exosomes were characterized by dynamic light scattering,

transmission electron microscopy and western blot. Coculture experiments were

performed to assess exosomal RNAs transferring from mesenchymal stem cells

(MSCs) to MM cells. The role of PSMA3-AS1 in PIs sensitivity was further evaluated

in vivo. To determine the prognostic significance of circulating exosomal PSMA3 and

PSMA3-AS1, a cohort of newly diagnosed MM patients was enrolled to study. Cox

regression models and Kaplan-Meier curves were used to analyze progression free

survival (PFS) and overall survival (OS).

Results: We identified that PSMA3 and PSMA3-AS1 in MSCs could be packaged

into exosomes and transferred to myeloma cells, thus promoted PIs resistance.

PSMA3-AS1 could form an RNA duplex with pre-PSMA3, which transcriptionally

promoted PSMA3 expression by increasing its stability. In xenograft models,

intravenously administered siPSMA3-AS1, was found to be effective in increasing

carfilzomib sensitivity. Moreover, plasma circulating exosomal PSMA3 and

PSMA3-AS1 derived from MM patients were significantly associated with PFS and

OS.

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Conclusion: This study suggested a unique role of exosomal PSMA3 and

PSMA3-AS1 in transmitting PIs resistance from MSCs to MM cells, through a novel

exosomal PSMA3-AS1/PSMA3 signaling pathway. Exosomal PSMA3 and

PSMA3-AS1 might act as promising therapeutic targets for PIs resistance and

prognostic predictors for clinical response.

Introduction

Multiple myeloma (MM) is a malignancy of the plasma cell characterized by

proliferation of plasma cell clones (1). The proteasome inhibitor bortezomib (BTZ)

has shown promise in the treatment of MM (2). However, its therapeutic activity was

severely impeded by BTZ resistance (3). Multifactorial mechanisms underlying PIs

resistance have been studied: genetic mutations, gene expression signatures, copy

number abnormalities and bone marrow microenvironment (4). Nevertheless, MM is a

multifaceted disease related with genetic, epigenetic and chromosomal alterations,

and the mechanisms underlying PIs resistance in MM remains elusive.

The proteasome is a 26S enzyme complex that consists of a 19S regulatory complex

and a core 20S catalytic complex. The 20S complex is composed of two rings of

seven α subunits and two rings of seven β subunits (5). PIs could slightly inhibit the

peptidyl glutamyl–like activity at the β1 subunit (PSMB6) and mainly target the

chymotrypsin-like activity of β5 subunit (PSMB5) (6, 7). The mutations in β5 subunit

or the increased expression of β5 subunit was detected in BTZ-resistant hematologic

tumor cell lines (8). However, the β5 mutations and overexpression are absent in

clinical samples derived from BTZ resistant patients (9, 10). Therefore, the β5 subunit

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

dysregulation may not be uniquely responsible for PIs resistance. It is conceivable

that other mechanisms could be associated with PIs resistance.

The bone marrow microenvironment activates many pathways leading to disease

progression. MSCs support tumor cell growth, metastasis and evasion of the immune

system (11). The interaction between MSCs and myeloma cells performs a critical

role in MM pathogenesis, progress and chemotherapy resistance (12, 13). These

findings indicate a need for developing novel drugs to counteract these cancer–stroma

interactions.

Exosomes act as key communicators between tumor microenvironment and cancer

cells (14, 15). The interaction between MSCs and MM cells plays a crucial role in

MM pathogenesis and drug resistance by exosomes (16, 17). Recently, lncRNAs have

been reported to exist in exosomes (18, 19). LncRNAs are transcribed from thousands

of loci in mammalian genomes and function in a wide range of biological processes

(20, 21). In cancers, lncRNAs are emerging important regulators in oncogenic

pathways (22, 23). However, the role of lncRNAs in the MM pathogenesis and

progression has not been fully elucidated. In particular, how exosomal lncRNAs

derived from the bone marrow microenvironment contribute to PIs resistance is

unexplored in MM.

Abundant bidirectional transcription of eukaryotic genes has been explored across

genomes (24). Emerging evidence suggests that antisense lncRNAs play vital roles in

regulating their sense transcripts partners, in particular, affecting the expression of

their associated protein coding genes (24, 25). However, we know little about the

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

precise function of natural antisense transcripts and the molecular mechanisms of

their biogenesis in MM.

This study explored how the MSCs-derived exosomal lncRNAs conferred PIs

resistance to MM cells and through which molecular mechanisms they elicited this

function. Our data underscored that exosomal lncPSMA3-AS1 was the key

determinant in mediating resistance to PIs by regulating the stability of PSMA3,

which encodes proteasome subunit α7. Upregulation of PSMA3 led to the α-subunit–

related increase in chymotrypsin-like proteasome activity. These combined

approaches targeting PMSA3 and PSMA3-AS1 in conjunction with PIs could enhance

therapeutic efficacy. Likewise, our findings suggest that the circulating exosomal

PSMA3 and PSMA3-AS1 from the plasma of MM patients could improve to identify

the newly diagnosed patients with poor outcomes.


Materials and methods
Patient Samples

Samples were taken from newly diagnosed MM patients who were enrolled to study.

The study was conducted in accordance with the Declaration of Helsinki. Informed

written consent was obtained from each subject or each subject's guardian. The human

investigations were performed after approval by an institutional review board and in

accordance with an assurance filed with. The detailed clinical features of these

patients were listed in Table S1. Progression-free survival (PFS) was defined as the

time between initiation of bortezomib therapy and the date of first evidence of

progressive. Patients who were progression-free at the time of analysis were censored

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

using the time between initial treatment and last follow-up. More details of patient

samples are available in supplementary data.

Plasmids construction

Full-length cDNA (PMSA3 or PSMA3-AS1) was synthesized and subcloned into the

EcoRI and BamHI sites of Lenti-CMV-puro vector, the constructs were verified by

DNA sequencing.

Exosomes purification

Exosomes secreted by cultured cell lines and MSCs were isolated using ExoQuick™

solution (System Biosciences). Exosomes in the plasma were isolated using

ExoQuick-LP™ solution (System Biosciences). The detailed procedures of exosome

isolation are available in supplementary data. The exosome pellet was isolated and the

protein content of the exosome suspension was analyzed by BCA protein assay kit

(Beyotime Biotechnology).

Dynamic Light Scattering analysis (DLS)

Dynamic Light Scattering analysis was performed to characterize and quantify the

particle size distribution of exosomes (Zetasizer Nano S90).

In vivo xenograft studies

The animal experiments were performed after acquired permission from the local

Ethical Committee according to the institutional guidelines for the use of laboratory

animals. More details of in vivo xenograft studies are available in supplementary data.

Statistical analysis

Survival analysis was performed to investigate the associations between the censored

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

outcomes and the clinical variables and the expression measures of PSMA3 and

PSMA3-AS1 using the R package survival. PFS and OS were the primary outcomes

of interest. The clinical stage (ISS) was treated as a three-level factor and thus coded

as two dummy variables (ISS2 and ISS3) to compare Stage I with Stages II and III.

All other clinical variables and the expression measures of PSMA3 and PSMA3-AS1

were numeric and were standardized to facilitate the comparison of hazards ratios

(HR).

We first used univariate Cox survival regression to separately analyze the predictor

variables. To illustrate the impact of the PSMA3 and PSMA3-AS1, we partitioned the

patients to two groups based on the medians of the PSMA3 and PSMA3-AS1

expressions, and estimated the survival curves of these two groups of patients using

the Kaplan-Meier method and compared their difference using the log-rank test. We

then used multivariate Cox survival regression to jointly fit all the clinical variables

with either PSMA3 or PSMA3-AS1, which adjusted for the potential confounding

effects of the clinical variables on PSMA3 or PSMA3-AS1. By fitting these Cox

models, we obtained the estimates of hazards ratios (HR), their 95% confidence

intervals (CIs), the corresponding p-values, and Harrell’s concordance (C-index). We

also validated the prognostic values of these models using the leave-one-out

cross-validation.

Pearson correlation analysis was employed to determine the correlation between the

expressions of PSMA3 or PSMA3-AS1. Student’s t test was performed to analyze the

difference of mean values between two groups for other continuous outcomes.

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Other detailed assays are available in supplementary data.

RESULTS

MSCs–derived exosomes were transferred to myeloma cells and conferred

proteasome inhibitors resistance to myeloma cells.

We first examined whether MSCs secreted exosomes. Primary r-MSCs obtained from

bortezomib-resistant patients, or s-MSCs obtained from bortezomib-sensitive patients

were cultured in serum-free medium for 48 hours. Then we isolated exosomes using

standard methods and confirmed their identity by transmission electron microscope

(Figure 1A). DLS analysis was performed to quantify the exosome size distribution

(Figure 1B). Western blot was used to confirm the expression of Flotillin-1 and

HSP70, which were generally used as exosome markers (Figure 1C).

Next we investigated whether MSCs–derived exosomes could be transmitted into

myeloma cells. Exosomes derived from MSCs were fluorescently labeled with

PKH67, and then cultured with MM.1S or U266. The ability of myeloma cells to take

MSCs–derived exosomes was confirmed using confocal microscope (Figure 1D,

Figure S1A) and flow cytometric assays (Figure 1E, Figure S1B). As the recipient

cells, myeloma cells showed the same uptake efficiency of exosomes from r-MSCs

and those from s-MSCs (Figure 1F). These results suggested that exosomes derived

from MSCs could be transmitted to myeloma cells, indicating a promising role in

regulating biologic functions of myeloma cells.

To further investigate the effect of MSCs-derived exosomes on the proteasome

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

inhibitors resistance, U266 or MM.1S cells were treated with or without different

amounts of exosomes in the presence of bortezomib or carfilzomib. R-MSCs-derived

exosomes significantly reduced proteasome inhibitors sensitivity (Figure 1G, Figure

S1C). However, no difference in the anti-proliferative activities of proteasome

inhibitors was observed between treatment with or without different amounts of

exosomes obtained from s-MSCs. These data demonstrated that exosomes derived

from r-MSCs, but not from s-MSCs reduced the proteasome inhibitors sensitivity in

MM cells.

Identifying mRNAs involved in bortezomib-resistant patients.

To explore the possible mechanisms of proteasome inhibitors resistance in multiple

myeloma, gene expression profiling of bortezomib-resistant patients (NC/PD) and

bortezomib-sensitive patients (CR/PR/MR) were analyzed using GEO database

(GSE9782). GSEA showed that the positive regulation of proteasome gene signatures

(the gene sets: BIOCARTA_PROTEASOME_PATHWAY,

WONG_PROTEASOME_GENE_MODULE, KEGG_PROTEASOME) were

significantly enriched in bortezomib-resistant samples (Figure 2A). A volcano plot

showed that three transcripts (PSMA3: fold change = 1.29/ p=0.0014, PSMA3-AS1:

fold change = 1.27/ p=0.000595, USP13: fold change = 1.257/ p=0.0013) were

significantly upregulated abundances in bortezomib-resistant samples compared to

bortezomib-sensitive samples (Figure 2B). Subsequently, we identified that PSMA3

and PSMA3-AS1, not USP13, were present in MSCs-derived exosomes (Figure 2C).

10

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Then we focused on PSMA3 and PSMA3-AS1 transcripts. PSMA3 and PSMA3-AS1

expression were higher in bortezomib-resistant samples compared with

bortezomib-sensitive samples (Figure 2D). In addition, Kaplan-Meier analysis showed

that high PSMA3 expression in CD138+ MM cells was associated with decreased PFS

(p = 0.0307) and poor OS (p = 0.0328) (Figure 2E). Cox proportional hazards

regression analysis further confirmed that high PSMA3 could act as an independent

prognostic factor for MM patients with bortezomib therapy in a multivariate analysis

(p = 0.0013, HR = 1.3104, 95%CI = 1.1113-1.545).

To investigate whether the PSMA3 content of MM was involved in facilitating

multiple myeloma progression, we analyzed the datasets obtained from the Oncomine

database to determine the PSMA3 expression alterations in CD138+ cells in

monoclonal gammopathy of undetermined significance (MGUS), smoldering

myeloma (SM), multiple myeloma (MM) and plasma cell leukemia (PCL). As shown

in Figure 2F, PSMA3 levels were increased in CD138+ cells from SM compared to

those from MGUS (26), and the PSMA3 expressions were upregulated in CD138+

cells from MM compared to those from MGUS (Figure 2G) (27). Moreover, the

PSMA3 levels appeared a progressive increase in MGUS, MM and PCL (Figure 2H)

(28). These analyses consistently suggested that the expression levels of PSMA3 in

CD138+ cells were correlated with MM progression.

Consistent with this previously published study, our clinical data showed that the

mRNA levels of PSMA3 and PSMA3-AS1 were upregulated in CD138+ MM cells

derived from bortezomib resistant patients relative to those from bortezomib sensitive

11

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

patients (Figure 2I). To further explore the mechanisms of proteasome inhibitors

resistance, MM cell lines with a 4-fold or greater resistance to bortezomib,

carfilzomib or ixazomib were developed. The proteasome inhibitors resistant

(bortezomib resistance, BR; carfilzomib resistance, CR; ixazomib resistance, IR)

models consistently displayed upregulation of PSMA3 and PSMA3-AS1 transcript

levels (Figure 2J, Figure S2). In addition, r-MSCs had increased expression of

PSMA3 and PSMA3-AS1 compared to s-MSCs (Figure 2K). Moreover, the

expressions of PSMA3 and PSMA3-AS1 in MSCs were positively correlated with

those in CD138+ myeloma cells (Figure 2L). These results suggested that high levels

of PSMA3 and PSMA3-AS1 were associated with proteasome inhibitors resistance of

myeloma cells.

Exosome-mediated transfer of PSMA3 and lncPSMA3-AS1 from MSCs to MM

cells contributed to proteasome inhibitors resistance

Next we investigated whether exosome-transferred PSMA3 and PSMA3-AS1 could

confer proteasome inhibitors resistance to MM cells. We found that PSMA3 and

PSMA3-AS1 levels statistically increased in MM cells incubated with

r-MSCs-derived exosomes. This change did not occur when MM cells were incubated

with s-MSCs-derived exosomes (Figure 3A, Figure S3A). Moreover, U266 or MM.1S

incubated with exosomes from r-MSCs but not those from s-MSCs showed an

increase in the chymotrypsin-like (ChT-L) activity of the proteasome (Figure S3B).

Treatment directly with r-MSCs-derived exosomes or s-MSCs-derived exosomes had

12

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

no effects on cell viability and apoptosis (Figure S3C, S3D).

To further evaluate the possibility that exosome-mediated transmission of PSMA3 and

PSMA3-AS1 from MSCs to MM cells contributed to proteasome inhibitors resistance,

we examined whether the effect could be reversed when PSMA3 or PSMA3-AS1

expression was downregulated in r-MSCs. As expected, the levels of PSMA3 or

PSMA3-AS1 were decreased in MM cells upon incubation with exosomes from

PSMA3 or PSMA3-AS1-knockdown r-MSCs compared to siRNA control cells

(Figure 3B, Figure S3E). Moreover, U266 or MM.1S incubated with exosomes from

r-MSCs displayed decreased proteasome inhibitors sensitivity, which could be

abolished by downregulating PSMA3 or PSMA3-AS1 in r-MSCs (Figure 3C, Figure

S3F). Consistent with the loss of function experiments, when PSMA3 or PSMA3-AS1

was upregulated in s-MSCs, levels of PSMA3 or PSMA3-AS1 were increased in MM

cells incubated with exosomes from PSMA3 or PSMA3-AS1-overexpression s-MSCs

compared to vector control cells (Figure 3B, Figure S3E). In concordance with

upregulation of PSMA3 and PSMA3-AS1, cells showed decreased proteasome

inhibitors sensitivity (Figure 3C, Figure S3F). Similar results were obtained when

directly knocking down PSMA3 or PSMA3-AS1 in U266 or MM.1S (Figure 3D,

Figure S3G). These data suggested that PSMA3 or PSMA3-AS1 level differences in

MSCs derived-exosome content could mediate PIs resistance in MM cells.

Previous study has showed that PSMA3 which encoding the constitutive proteasome

component, α7 subunit, performed an important role in proteasome formation and

function (29). To further determine the role of PSMA3 and PSMA3-AS1 in

13

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

proteasome inhibitors sensitivity in MM cells, we examine whether PSMA3 and

PSMA3-AS1 levels were associated with proteasome activity. As expected,

knockdown of PSMA3 or PSMA3-AS1 caused a decrease in the chymotrypsin-like

(ChT-L) activity of the proteasome in U266 or MM.1S, whereas upregulated either

PSMA3 or PSMA3-AS1 enhanced the ChT-L proteasome activity (Figure 3E, Figure

S3H). These findings demonstrated that knockdown of either PSMA3 or PSMA3-AS1

increased the proteasome inhibitors sensitivity. Consistent with this finding,

overexpressing either PSMA3 or PSMA3-AS1 reduced the proteasome inhibitors

sensitivity. Taken together, these data indicated that the MSCs-derived exosomal

PSMA3 and PSMA3-AS1 conferred proteasome inhibitors resistance to the MM cells.

Higher levels of PSMA3 and PSMA3-AS1 were less sensitive to the proteasome

inhibitors, which produced a greater increase in cell proliferation and

chymotrypsin-like proteasome activity.

Next, we determined whether downregulation of PSMA3 and PSMA3-AS1 would

lead to restoration of PIs sensitivity in PIs resistance myeloma cells. Either

PSMA3-siRNA–dependent prevention or PSMA3-AS1-siRNA–dependent prevention

after PIs exposure was accompanied by both a significantly lower cell proliferation

(Figure 3G), higher fraction of apoptotic cells (Figure 3H) as well as decreased ChT-L

proteasome activity (Figure 3F). Collectively, these results further underscored the

involvement of PSMA3 and PSMA3-AS1 overexpression in conferring PIs resistance

and provided a targeted strategy to reverse PIs resistance.

14

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

PSMA3-AS1 reduced proteasome inhibitors sensitivity by regulating PSMA3

expression in MM cells.

In order to identify lncRNA PSMA3-AS1 involved in PIs resistance in MM, we

characterized the PSMA3-AS1. RNA fluorescence in situ hybridization (FISH) and

nuclear and cytoplasmic fractions assays indicated that PSMA3-AS1 existed both in

the nucleus and in the cytoplasm (Figure 4A, 4B). Rapid amplification of cDNA ends

(RACE) was performed for directional sequencing of 5′ and 3′ ends and two splice

variants, 2531 nt and 2440 nt were identified (Figure 4C). We found the presence of a

poly-A tail, indicating that PSMA3-AS1 is a fully processed transcript of RNA

polymerase II. Coding Potential Calculator (CPC, http://cpc.cbi.pku.edu.cn/) (30) and

Coding Potential Assessment Tool (CPAT, http://lilab.research.bcm.edu/cpat/) (31)

analyses displayed that PSMA3-AS1 has no coding potentiality (Table S4, Table S5).

Northern blotting assay further confirmed the existence of full length PSMA3-AS1 in

MM.1S (Figure 4D).

It is reported that the antisense transcripts can commonly regulate the sense transcripts

expressions in two ways (32). In concordant way, the antisense transcripts have

positive effects on sense transcripts (33). Conversely, the level of the sense RNA, or

corresponding protein level is negatively regulated by the antisense transcripts in

discordant way (34). To explore the biology of PSMA3-AS1 and consider its potential

significance, the correlation between PSMA3-AS1 and PSMA3 was examined in MM

cells derived from 57 MM patients by Pearson correlation analysis. The results

showed that PSMA3-AS1was positively correlated with PSMA3 in MM cells (Figure

15

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

4E).

To examine whether the PSMA3-AS1 transcript regulated the PSMA3 expression, we

transiently transfected siRNA PSMA3-AS1 to U266 or MM.1S. We found that

knockdown PSMA3-AS1 significantly decreased PSMA3 level in U266 or MM.1S

(Figure 4F, Figure S4A).

Given that PSMA3-AS1 knockdown decreased the proteasome activity and

subsequently reduced the cell viability (Figure 3D, 3E, Figure S3G, S3F), we sought

to determine whether reconstituting the expression of PSMA3 in PSMA3-AS1

downregulated cells could reverse the altered biological activity. We found that

restoring the expression of PSMA3 in PSMA3-AS1 knockdown cells significantly

abrogated the reduced ChT-L activity and the increased proteasome inhibitors

sensitivity conferred by PSMA3-AS1 downregulation in MM cells (Figure 4G, 4H,

Figure S4B, S4C). These data demonstrated that PSMA3-AS1 had effects on

proteasome activity and proteasome inhibitors sensitivity by regulating PSMA3

expression in MM cells.

PSMA3-AS1 formed an RNA duplex with PSMA3-AS1 pre-mRNA and increased

its stability.

Antisense transcripts commonly interact with their sense transcripts in a

double-stranded RNA structures form, which may potentially regulates the transport,

splicing and stability of the sense transcripts (24). PSMA3 and PSMA3-AS1 genes

are overlap and both located at chromosome 14p23.1. They are transcribed in opposite

16

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

directions. PSMA3-AS1 overlaps the 2029 nucleotides within the 7th intron of

PSMA3 (Figure S5A). To verify the existence of RNA duplex structure between

PSMA3-AS1 and pre-PSMA3, RNase protection assay was performed on RNAs from

U266 and MM.1S. RNase A digested the single-stranded RNA and the remaining

double-stranded RNA was protected from degradation. According to the PCR assays

using indicated primers, the overlapping part of both transcripts existed, which

suggesting that PSMA3-AS1 and pre-PSMA3 indeed form a RNA duplex (Figure

S5B). RNA-RNA duplex could change the secondary or tertiary structure of RNA and

protect RNA from RNase degradation, thereby increase its stability (33, 35). To

investigate whether the duplex form between PSMA3-AS1 and pre-PSMA3 affected

the PSMA3 mRNA stability, we assessed the stability of PSMA3 transcripts by

blocking new RNA synthesis with α-amanitin (an RNA polymerase II inhibitor) and

measuring the loss of PSMA3, PSMA3-AS1, GAPDH and 18s RNA over a 24h

period (Figure S5C). The results showed that knockdown PSMA3-AS1 decreased

stability of PSMA3. Conversely, PSMA3-AS1 overexpression increased stability of

PSMA3 (Figure S5D). Together, these results demonstrated that PSMA3-AS1 formed

an RNA duplex with PSMA3-AS1 pre-mRNA, which led to enhance the stability of

PSMA3 by reducing its decay.

Targeting PSMA3-AS1 increased proteasome inhibitors sensitivity in vivo.

To evaluate the therapeutic potential of PSMA3-AS1 in MM in vivo, we established

bioluminescent MM models (U266-luc), which recapitulates the bone

17

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

pathophysiology and the clinical sequelae observed in myeloma patients (36) (Figure

5A). The mRNA levels of PSMA3-AS1 and PSMA3 in the mouse CD138+ cells were

performed by qPCR to confirm the injected siRNA efficiency in vivo (Figure 5C).

Bioluminescent imaging revealed that therapeutic PSMA3-AS1 siRNA efficiently

increased the sensitivity of U266 xenografts to carfilzomib treatment (Figure 5B).

Moreover, the combination treatment with PSMA3-AS1 siRNA and carfilzomib

significantly prolonged the overall survival compared to carfilzomib combination

with siRNA control or siPSMA3-AS1-treated alone (Figure 5D). No significant

weight loss or unexpected deaths was found during treatment. Collectively,

downregulation of PSMA3-AS1 in vivo significantly enhanced the sensitivity of MM

xenografts to carfilzomib. These data revealed that PSMA3-AS1 was essential for

proteasome inhibitors resistance in MM cells.

Prognostic role of circulating exosomal PSMA3 and PSMA3-AS1 in multiple

myeloma

Several cell types, including cancer cells secrete exosomes. Exosomes have been

reported to be isolated from the peripheral blood and act as powerful markers for

disease progression in cancer (37, 38). To determine the prognostic significance of

circulating exosomal PSMA3 and PSMA3-AS1 in MM, we isolated and characterized

exosomes from the plasma of MM patients. A cohort of 57 newly diagnosed MM

patients was uniformly treated and followed. QPCR assays were performed to analyze

circulating exosomal RNAs derived from plasma of MM patients.

18

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

First, we isolated plasma circulating exosomes from MM patients and confirmed them

by transmission electron microscope (Figure 6A) and dynamic light scattering assays

(Figure 6B). Similar numbers of exosomes were isolated from bortezomib resistant

patients and bortezomib sensitive patients (Figure 6C). By comparing PSMA3 and

PSMA3-AS1 levels in circulating exosomes and the corresponding exosome-depleted

plasma, we found that circulating PSMA3 and PSMA3-AS1 predominantly existed in

exosomes (Figure 6D). Moreover, expressions of PSMA3 and PSMA3-AS1 in

circulating exosomes were positively correlated with that in CD138+ myeloma cells

(Figure 6E).

The results from the univariate and multivariate Cox survival regressions were present

in Table S6. We can see that Exosmic PSMA3 and PSMA3-AS1 were significantly

associated with both PFS and OS in the univariate and multivariate analyses (Table S6,

Figure 6F, 6G). We compared the C-indices of the models fitting the ISS alone, ISS

with PSMA3, and ISS with PSMA3-AS1 to show the predictive value of the PSMA3

and PSMA3-AS1 expressions (Table S7). In order to validate the prognostic values of

PSMA3 and PSMA3-AS1, the leave-one-out cross-validation was performed (Table

S7). The results showed that both exosomal PSMA3 and PSMA3-AS1 were

significant predictive biomarkers on PFS and OS. These data indicated that the

exosmal PSMA3 or PSMA3-AS1 could be crucial in identifying worse prognosis in

newly diagnosed MM patients. Therefore, it is advisable to evaluate PSMA3 and

PSMA3-AS1 expression in MM to identify patients who might benefit from

bortezomib therapy before deciding on a course of treatment.

19

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Discussion

Resistance to the PIs is a major clinical problem in MM and the mechanisms have not

been completely revealed. The bone marrow microenvironment could promote the

survival and drug resistance of myeloma cells. It has been proposed that MSCs

contribute to MM microenvironment, where they can promote or suppress MM

progression (11, 39). We aimed to find a way to interfere these networks to stop tumor

growth and drug resistance.

Many types of cells could release exosomes (70–120 nm) into the extracellular

environment (38, 40). Exosomes are involved in intercellular communication, and in

this study we investigated how the transfer of exosomal PMSA3 and PSMA3-AS1

from MSCs to MM cells affected proteasome inhibitors resistance (Figure 6H).

Proteasome inhibitors resistance is a vital impediment to the clinical treatment.

Numerous underlying mechanisms have been identified (41-43), but the primary

cause is still unknown. Initial studies in hematologic tumor cell line models

demonstrated that overexpression of the β5 proteasome subunit or mutations in the β5

subunit’s bortezomib binding pocket were implicated in bortezomib resistance (8, 44).

However, the β5 mutations and overexpression are absent in clinical specimen

obtained from BTZ resistant patients (9, 10). To further elucidate mechanisms of PIs

resistance, we retrieved a database containing gene expression profile of 169

myeloma cases with clinical response and disease prognosis (45). The analysis of this

dataset showed that the mRNA levels of PSMA3 and PSMA3-AS1 in CD138+ cells

20

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

are upregulated in bortezomib-resistant patients. Moreover, increased PSMA3 and

PSMA3-AS1 mRNA expression correlated with proteasome inhibitors resistance and

poor outcome for myeloma patients treated with bortezomib. Further analyses of

Oncomine data showed that the PSMA3 levels appeared a progressive increase in

MGUS, SM, MM and PCL (26, 28). Similarly, our PIs resistant models (U266BR,

U266CR, U266IR, MM.1SBR, MM.1SCR, MM.1SIR) consistently displayed

up-regulation of PSMA3 and PSMA3-AS1 expression. These results suggested that

the PIs insensitivity in MM was due to up-regulated PSMA3 and PSMA3-AS1.

LncRNAs are extensively involved in cellular processes, including cell cycle,

apoptosis and immunity (23, 46). LncRNAs fulfill their functions through multiple

mechanisms, such as scaffolding of nuclear or cytoplasmic complexes, regulation of

gene expression and pairing with DNA or RNAs. Recently, several protein-coding

mRNAs have been reported to have natural antisense transcript partners, most of

which are noncoding RNAs (20, 47). Natural antisense lncRNA transcripts could

serve as therapeutic targets through regulating the sense gene expression (25, 48). In

this study, we identified a natural antisense transcript, PSMA3-AS1, arises from

intergenic regions of the PSMA3 gene. Our data indicated that as a pair of sense

protein-coding / antisense non-coding transcripts, PSMA3-AS1 could form a duplex

structure with PSMA3 pre-mRNA, thereby increasing its stability by reducing its

decay. And by enhancing the stability of PSMA3 pre-mRNA, PSMA3-AS1

upregulated the PSMA3 expression. In addition, downregulation of PSMA3-AS1

decreased PSMA3 expression, and subsequently reducing the chymotrypsin-like

21

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

proteasome activity and increasing the PIs sensitivity in myeloma cells. Importantly,

therapeutic lncPSMA3-AS1 siRNA efficiently increased the sensitivity of U266

xenografts to carfilzomib treatment, suggesting the significance of lncPSMA3-AS1 in

MM. Collectively, our data showed that disturbance of an antisense RNA

PSMA3-AS1 could alter the expression of sense mRNA PSMA3, suggesting that

antisense transcript PSMA3-AS1 contributed to the regulation of PSMA3 in MM cells.

These results suggested that many lncRNAs might exert their functions by RNA-RNA

direct interactions and that widely studying these interactions may further explore

lncRNA function and mechanism.

Exosomal RNAs have been reported to contribute to some aspects of oncology,

including tumorigenesis, metastasis and chemoresistance (15, 38). Recent evidence

suggests that more than twenty percent of exosomal RNAs are lncRNAs (49). Our in

vitro data indicated that lncPSMA3-AS1 could be packaged into exosomes and

transmitted to myeloma cells, conferred PIs resistance to MM cells by increasing the

PSMA3 expression.

Circulating exosomal RNAs have been implicated to act as potential non-invasive

molecular predictors of prognosis and drug response (50). However, the discovery of

specific exosomal RNAs as disease-specific biomarkers depends on validated

methods for sample isolation and analyses. Here, we identified circulating exosomal

PSMA3 and PSMA3-AS1 as clinically relevant biomarkers using standardized

techniques for sample collection and exosomal RNAs measurement. Another

challenge is that RNA copy numbers are generally low in circulating exosomes, which

22

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

makes it difficult to obtain sufficient RNA for analyses (50). In this study, we did

observe that PSMA3 and PSMA3-AS1 were relatively enriched in exosomes derived

from the plasma of MM patients by qPCR. Collectively, we identified that circulating

exosomal PSMA3 and PSMA3-AS1 derived from the plasma of MM patients were

associated with poor outcomes in regards to PFS and OS. The levels of exosomal

PSMA3 and PSMA3-AS1 could serve as prognostic factors for MM in a multivariate

Cox regression analysis. Our study provided evidence of the association between

circulating exosomal lncRNAs and outcomes in patients with newly diagnosed MM.

Here, we revealed that the informative lncRNAs derived from MSCs exosomes

involved in the communication between the MSCs and myeloma cells, thus resulted

in PIs resistance in MM. Our findings confirmed that PIs resistance was conferred by

increased expression of PMSA3 and PSMA3-AS1. The data also suggested that low

expression of PSMA3 and PSMA3-AS1 in pre-therapy MM exhibited a significant

improvement in prognosis after receiving bortezomib, whereas patients with high

PSMA3 and PSMA3-AS1 expressions were correlated with a poor bortezomib

response.

References

1.Kyle RA, Gertz MA, Witzig TE, Lust JA, Lacy MQ, Dispenzieri A, et al. Review of 1027

patients with newly diagnosed multiple myeloma. Mayo Clin Proc 2003;78: 21-33.

2.Hideshima T, Chauhan D, Richardson P, Anderson KC Identification and validation of novel

therapeutic targets for multiple myeloma. J Clin Oncol 2005;23: 6345-6350.

23

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

3.Ahn JS, Jung SH, Yang DH, Bae SY, Kim YK, Kim HJ, et al. Patterns of relapse or

progression after bortezomib-based salvage therapy in patients with relapsed/refractory

multiple myeloma. Clin Lymphoma Myeloma Leuk 2014;14: 389-394.

4.Nikesitch N, Ling SC Molecular mechanisms in multiple myeloma drug resistance. J Clin

Pathol 2016;69: 97-101.

5.Chen P, Hochstrasser M Autocatalytic subunit processing couples active site formation in

the 20S proteasome to completion of assembly. Cell 1996;86: 961-972.

6.Kisselev AF, Callard A, Goldberg AL Importance of the different proteolytic sites of the

proteasome and the efficacy of inhibitors varies with the protein substrate. The Journal of

biological chemistry 2006;281: 8582-8590.

7.Crawford LJ, Walker B, Ovaa H, Chauhan D, Anderson KC, Morris TC, et al. Comparative

selectivity and specificity of the proteasome inhibitors BzLLLCOCHO, PS-341, and MG-132.

Cancer research 2006;66: 6379-6386.

8.Oerlemans R, Franke NE, Assaraf YG, Cloos J, van Zantwijk I, Berkers CR, et al. Molecular

basis of bortezomib resistance: proteasome subunit beta5 (PSMB5) gene mutation and

overexpression of PSMB5 protein. Blood 2008;112: 2489-2499.

9.Franke NE, Niewerth D, Assaraf YG, van Meerloo J, Vojtekova K, van Zantwijk CH, et al.

Impaired bortezomib binding to mutant beta5 subunit of the proteasome is the underlying basis

for bortezomib resistance in leukemia cells. Leukemia 2012;26: 757-768.

10.Lichter DI, Danaee H, Pickard MD, Tayber O, Sintchak M, Shi H, et al. Sequence analysis

of beta-subunit genes of the 20S proteasome in patients with relapsed multiple myeloma

treated with bortezomib or dexamethasone. Blood 2012;120: 4513-4516.

24

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

11.Kuehl WM, Bergsagel PL Molecular pathogenesis of multiple myeloma and its

premalignant precursor. The Journal of clinical investigation 2012;122: 3456-3463.

12.McDonald MM, Fairfield H, Falank C, Reagan MR Adipose, Bone, and Myeloma:

Contributions from the Microenvironment. Calcif Tissue Int 2017;100: 433-448.

13.Tassone P, Neri P, Carrasco DR, Burger R, Goldmacher VS, Fram R, et al. A clinically

relevant SCID-hu in vivo model of human multiple myeloma. Blood 2005;106: 713-716.

14.Hsu YL, Hung JY, Chang WA, Lin YS, Pan YC, Tsai PH, et al. Hypoxic lung

cancer-secreted exosomal miR-23a increased angiogenesis and vascular permeability by

targeting prolyl hydroxylase and tight junction protein ZO-1. Oncogene 2017.

15.Shao H, Chung J, Lee K, Balaj L, Min C, Carter BS, et al. Chip-based analysis of exosomal

mRNA mediating drug resistance in glioblastoma. Nature communications 2015;6: 6999.

16.Roccaro AM, Sacco A, Maiso P, Azab AK, Tai YT, Reagan M, et al. BM mesenchymal

stromal cell-derived exosomes facilitate multiple myeloma progression. The Journal of clinical

investigation 2013;123: 1542-1555.

17.Wang J, Hendrix A, Hernot S, Lemaire M, De Bruyne E, Van Valckenborgh E, et al. Bone

marrow stromal cell-derived exosomes as communicators in drug resistance in multiple

myeloma cells. Blood 2014;124: 555-566.

18.Qu L, Ding J, Chen C, Wu ZJ, Liu B, Gao Y, et al. Exosome-Transmitted lncARSR

Promotes Sunitinib Resistance in Renal Cancer by Acting as a Competing Endogenous RNA.

Cancer cell 2016;29: 653-668.

19.Li Q, Shao Y, Zhang X, Zheng T, Miao M, Qin L, et al. Plasma long noncoding RNA

protected by exosomes as a potential stable biomarker for gastric cancer. Tumour biology : the

25

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

journal of the International Society for Oncodevelopmental Biology and Medicine 2015;36:

2007-2012.

20.Wang P, Xue Y, Han Y, Lin L, Wu C, Xu S, et al. The STAT3-binding long noncoding RNA

lnc-DC controls human dendritic cell differentiation. Science 2014;344: 310-313.

21.Scarola M, Comisso E, Pascolo R, Chiaradia R, Marion RM, Schneider C, et al. Epigenetic

silencing of Oct4 by a complex containing SUV39H1 and Oct4 pseudogene lncRNA. Nature

communications 2015;6: 7631.

22.Liu B, Sun L, Liu Q, Gong C, Yao Y, Lv X, et al. A cytoplasmic NF-kappaB interacting long

noncoding RNA blocks IkappaB phosphorylation and suppresses breast cancer metastasis.

Cancer cell 2015;27: 370-381.

23.Wang Y, He L, Du Y, Zhu P, Huang G, Luo J, et al. The long noncoding RNA lncTCF7

promotes self-renewal of human liver cancer stem cells through activation of Wnt signaling.

Cell Stem Cell 2015;16: 413-425.

24.Faghihi MA, Wahlestedt C Regulatory roles of natural antisense transcripts. Nat Rev Mol

Cell Biol 2009;10: 637-643.

25.Carrieri C, Cimatti L, Biagioli M, Beugnet A, Zucchelli S, Fedele S, et al. Long non-coding

antisense RNA controls Uchl1 translation through an embedded SINEB2 repeat. Nature

2012;491: 454-457.

26.Zhan F, Barlogie B, Arzoumanian V, Huang Y, Williams DR, Hollmig K, et al.

Gene-expression signature of benign monoclonal gammopathy evident in multiple myeloma is

linked to good prognosis. Blood 2007;109: 1692-1700.

27.Zhan F, Hardin J, Kordsmeier B, Bumm K, Zheng M, Tian E, et al. Global gene expression

26

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

profiling of multiple myeloma, monoclonal gammopathy of undetermined significance, and

normal bone marrow plasma cells. Blood 2002;99: 1745-1757.

28.Agnelli L, Mosca L, Fabris S, Lionetti M, Andronache A, Kwee I, et al. A SNP microarray

and FISH-based procedure to detect allelic imbalances in multiple myeloma: an integrated

genomics approach reveals a wide gene dosage effect. Genes Chromosomes Cancer

2009;48: 603-614.

29.Brehm A, Liu Y, Sheikh A, Marrero B, Omoyinmi E, Zhou Q, et al. Additive loss-of-function

proteasome subunit mutations in CANDLE/PRAAS patients promote type I IFN production.

The Journal of clinical investigation 2015;125: 4196-4211.

30.Kong L, Zhang Y, Ye ZQ, Liu XQ, Zhao SQ, Wei L, et al. CPC: assess the protein-coding

potential of transcripts using sequence features and support vector machine. Nucleic Acids

Res 2007;35: W345-349.

31.Wang L, Park HJ, Dasari S, Wang S, Kocher JP, Li W CPAT: Coding-Potential Assessment

Tool using an alignment-free logistic regression model. Nucleic Acids Res 2013;41: e74.

32.Katayama S, Tomaru Y, Kasukawa T, Waki K, Nakanishi M, Nakamura M, et al. Antisense

transcription in the mammalian transcriptome. Science 2005;309: 1564-1566.

33.Faghihi MA, Modarresi F, Khalil AM, Wood DE, Sahagan BG, Morgan TE, et al. Expression

of a noncoding RNA is elevated in Alzheimer's disease and drives rapid feed-forward

regulation of beta-secretase. Nat Med 2008;14: 723-730.

34.Wahlestedt C Natural antisense and noncoding RNA transcripts as potential drug targets.

Drug Discov Today 2006;11: 503-508.

35.Su WY, Li JT, Cui Y, Hong J, Du W, Wang YC, et al. Bidirectional regulation between

27

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

WDR83 and its natural antisense transcript DHPS in gastric cancer. Cell Res 2012;22:

1374-1389.

36.Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Shringarpure R, Akiyama M, et al.

Inhibition of the insulin-like growth factor receptor-1 tyrosine kinase activity as a therapeutic

strategy for multiple myeloma, other hematologic malignancies, and solid tumors. Cancer cell

2004;5: 221-230.

37.Manier S, Liu CJ, Avet-Loiseau H, Park J, Shi J, Campigotto F, et al. Prognostic role of

circulating exosomal miRNAs in multiple myeloma. Blood 2017;129: 2429-2436.

38.Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, et al. Cancer-secreted miR-105

destroys vascular endothelial barriers to promote metastasis. Cancer cell 2014;25: 501-515.

39.Reagan MR, Ghobrial IM Multiple myeloma mesenchymal stem cells: characterization,

origin, and tumor-promoting effects. Clinical cancer research : an official journal of the

American Association for Cancer Research 2012;18: 342-349.

40.Boelens MC, Wu TJ, Nabet BY, Xu B, Qiu Y, Yoon T, et al. Exosome transfer from stromal

to breast cancer cells regulates therapy resistance pathways. Cell 2014;159: 499-513.

41.Tomlin FM, Gerling-Driessen UIM, Liu YC, Flynn RA, Vangala JR, Lentz CS, et al. Inhibition

of NGLY1 Inactivates the Transcription Factor Nrf1 and Potentiates Proteasome Inhibitor

Cytotoxicity. ACS Cent Sci 2017;3: 1143-1155.

42.Bianchi G, Oliva L, Cascio P, Pengo N, Fontana F, Cerruti F, et al. The proteasome load

versus capacity balance determines apoptotic sensitivity of multiple myeloma cells to

proteasome inhibition. Blood 2009;113: 3040-3049.

43.Shabaneh TB, Downey SL, Goddard AL, Screen M, Lucas MM, Eastman A, et al. Molecular

28

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

basis of differential sensitivity of myeloma cells to clinically relevant bolus treatment with

bortezomib. PloS one 2013;8: e56132.

44.Lu S, Yang J, Chen Z, Gong S, Zhou H, Xu X, et al. Different mutants of PSMB5 confer

varying bortezomib resistance in T lymphoblastic lymphoma/leukemia cells derived from the

Jurkat cell line. Exp Hematol 2009;37: 831-837.

45.Mulligan G, Mitsiades C, Bryant B, Zhan F, Chng WJ, Roels S, et al. Gene expression

profiling and correlation with outcome in clinical trials of the proteasome inhibitor bortezomib.

Blood 2007;109: 3177-3188.

46.Hudson WH, Pickard MR, de Vera IM, Kuiper EG, Mourtada-Maarabouni M, Conn GL, et al.

Conserved sequence-specific lincRNA-steroid receptor interactions drive transcriptional

repression and direct cell fate. Nature communications 2014;5: 5395.

47.He Y, Vogelstein B, Velculescu VE, Papadopoulos N, Kinzler KW The antisense

transcriptomes of human cells. Science 2008;322: 1855-1857.

48.Saldana-Meyer R, Gonzalez-Buendia E, Guerrero G, Narendra V, Bonasio R,

Recillas-Targa F, et al. CTCF regulates the human p53 gene through direct interaction with its

natural antisense transcript, Wrap53. Genes & development 2014;28: 723-734.

49.Huang X, Yuan T, Tschannen M, Sun Z, Jacob H, Du M, et al. Characterization of human

plasma-derived exosomal RNAs by deep sequencing. BMC genomics 2013;14: 319.

50.Bardelli A, Pantel K Liquid Biopsies, What We Do Not Know (Yet). Cancer cell 2017;31:

172-179.

Figure legends

Figure 1. MSCs derived exosomes were transferred to myeloma cells and


29

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

conferred proteasome inhibitors resistance to myeloma cells. (A) Representative

TEM imaging of exosomes derived from r-MSCs and s-MSCs (Bar = 100 nm). (B)

Exosomes isolated from r-MSCs and s-MSCs were measured by Dynamic Light

Scattering. (C) Representative western blot of HSP70 and flotillin-1 proteins in

r-MSCs-derived exosomes and s-MSCs-derived exosomes. (D) Myeloma cells were

cultured in the presence of MSCs–derived PKH67-labeled exosomes for 24hours.

Exosome uptakes by myeloma cells were shown using a confocal microscope

(original magnification, ×400). Myeloma cells were stained using DAPI (nuclei) and

594 conjugated anti-actin antibody. Scale bars, 10 μm. (E) Flow cytometric analysis

of U266 after incubation with fluorescently labeled exosomes. FL1 fluorescence

indicates exosome uptake. (F) Flow cytometric analysis of myeloma cells after

incubation with fluorescently labeled exosomes from r-MSCs and s-MSCs for

indicated time. (G) U266 and MM.1S cells were planted in 96-well plates with or

without different amounts of exosomes (5 μg, 20 μg, 40 μg) and bortezomib or

carfilzomib for 72h. The cell viability was measured using CCK-8. Error bars

represent the mean ± SD of 3 independent experiments, **, P < 0.01.

Figure 2. Identifying mRNAs involved in bortezomib-resistant patients. (A)

Specific baseline gene expression signatures are associated with proteasome activity.

Representative GSEA plots illustrating: The enrichment of genes involved in positive

regulation of proteasome among the transcripts in bortezomib-resistant patients. FDR,

false discovery rate; NES, normalized enrichment score. (B) Volcano plot showing the

30

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

gene expressions changes in the proteasome activity gene sets between

bortezomib-resistant samples and bortezomib-sensitive samples. PSMA3,

PSMA3-AS1 and USP13 which had a significantly altered expression were

highlighted in red. Vertical and horizontal dashed lines indicated cutoff values (p

value of 0.05 and absolute logarithmic fold change of 1.25). (C) 1/Ct values from

qPCR analysis to determine the loading efficiency of transcripts. The mean 1/Ct and

standard deviation of the three independent experiments are shown. (D) Analysis of

PSMA3 and PSMA3-AS1 expression in bortezomib-resistant MM (NC/PD) and

bortezomib-sensitive MM (CR/PR/MR) using the GEO database (GSE9782). (E)

Kaplan-Meier analysis of PFS and OS in the high and low PSMA3 groups according

to the median PSMA3 level in CD138+ MM cells. (F) (G) (H) PSMA3 mRNA

expression in CD138+ cells from MGUS, SM, MM or PCL was analyzed by using

Oncomine database. Data were presented as Log2 median-centered intensity. MGUS:

monoclonal gammopathy of undetermined significance, SM: smoldering myeloma,

MM: multiple myeloma, PCL: plasma cell leukemia. (I) Analysis of PSMA3 and

PSMA3-AS1 expression in bortezomib-resistant MM (SC/PD) and

bortezomib-sensitive MM (sCR/CR/VGPR/PR/MR) using our clinical data. RR:

relapsed/refractory patients, ND: newly diagnosed patients. (J) PSMA3 and

PSMA3-AS1 expression were determined in U266 drug-naïve, bortezomib-resistant

(B20R), carfilzomib-resistant (C40R), ixazomib-resistan (I20R) cells using qPCR.

Proteasome inhibitor resistant cells had increased expression of PSMA3 and

PSMA3-AS. (K) Analysis of PSMA3 and PSMA3-AS1 expression in r-MSCs and

31

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

s-MSCs. (L) Pearson correlation analysis between PSMA3 and PSMA3-AS1 levels in

CD138+ myeloma cells and that in MSCs. Error bars represent the mean ± SD of 3

independent experiments, *, P < 0.05, **, P < 0.01 and ***, P < 0.001.

Figure 3. Exosome-mediated transfer of PSMA3 and PSMA3-AS1 from MSCs to

MM cells contributed to proteasome inhibitors resistance. (A) QPCR analysis of

PSMA3 and PSMA3-AS1 expression in U266 after incubation with indicated

exosomes. R-exo: R-MSCs-derived exosomes; S-exo: S-MSCs-derived exosomes. (B)

QPCR analysis of PSMA3 in U266 after incubation with indicated exosomes.

oePSMA3: PSMA3-overexpressing; oePSMA3-AS1: PSMA3-AS1-overexpressing;

oeVec: empty vector. (C) MSCs were transfected with indicated siRNAs or

overexpressing vectors for 24 hours, and exosomes were isolated. These exosomes

were then treated with RNase to remove unincorporated RNAs. Cell viability of U266

was assessed 48 hours after incubation with or without indicated exosomes and

bortezomib using CCK-8 assay. (D) U266 cells were transfected with PSMA3 siRNA,

PSMA3-AS1 siRNA, control siRNA, PSMA3-overexpressing vector,

PSMA3-AS1-overexpressing vector or empty vector and treated with or without

bortezomib. The cell viability was measured using CCK-8. (E) U266 cells were

transfected with PSMA3 siRNA, PSMA3-AS1 siRNA, control siRNA,

PSMA3-overexpressing vector, PSMA3-AS1-overexpressing vector or empty vector.

The cell numbers were counted and a proteasome activity assay was done. Fold

changes of activity against no-treatment control was calculated. (F) (G) (H) After

32

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

transfection of with PSMA3 siRNA, PSMA3-AS1 siRNA, control siRNA, PIs

resistance cells were incubated (24-48 hours) with bortezomib, carfilzomib or

ixazomib, respectively. The proteasome activity (F) and cell viability (G) were

measured as described previously. The apoptosis was measured by Annexin V

/7-AAD apoptosis detection kit (H). Error bars represent the mean ± SD of 3

independent experiments, *, P < 0.05, **, P < 0.01 and ***, P < 0.001.

Figure 4. PSMA3-AS1 reduced proteasome inhibitors sensitivity by regulating

PSMA3 expression in MM cells. Characterization of lncRNA PSMA3-AS1. The

nuclear and cytoplasmic location of PSMA3-AS1 in MSCs. (A) RNA FISH assay of

PSMA3-AS1 in U266, MM.1S and MSCs. Scale bars, 10 μm. (B) QPCR detection of

PSMA3-AS1 in the cytoplasmic (C) and nuclear (N) fractions. GAPDH served as an

internal normalization control. (C) Gel electrophoresis of nested PCR products from

5'-RACE and 3'-RACE. (D) Northern blot analysis with in vitro–transcribed

strand-specific RNA probes for PSMA3-AS1 in MM.1S. β-actin serves as a loading

control. (E) Correlation between PSMA3 and PSMA3-AS1 in CD138+ myeloma cells

from myeloma patients. r = 0.538, p < 0.001. (F) U266 or MM.1S were transiently

transfected with PSMA3-AS1siRNA or a control siRNA. QPCR was performed to

determine gene levels, with GAPDH used as an internal normalization control.

PSMA3 protein expression was determined by western blot. (G) U266 cells were

transfected with PSMA3-AS1 siRNA, control siRNA with or without

PSMA3-overexpressing vector or empty vector. The cell numbers were counted and

33

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

the proteasome activity assay were done. Fold changes of activity against

no-treatment control was calculated. (H) U266 cells were treated with or without

bortezomib and transfected with PSMA3-AS1 siRNA or control siRNA, with or

without PSMA3-overexpressing vector or empty vector. The cell viability was

measured using CCK-8. Error bars represent the mean ± SD of 3 independent

experiments, *, P < 0.05, **, P < 0.01 and ***, P < 0.001

Figure 5. Targeting PSMA3-AS1 increased proteasome inhibitors sensitivity in

vivo. (A) NSG mice were transplanted with U266-luc+cells and the combined

chemotherapy regimens were started on day 15 post xeno-transplantation as indicated

in the upper panel. (B) Representative whole-body bioluminescence images of NSG

mice. Therapy of carfilzomib in combination with PSMA3-AS1 siRNA or siRNA

control or PSMA3-AS1 siRNA alone in vivo was performed. Tumor development was

monitored by noninvasive bioluminescence imaging. (n = 5/group). Error bars

represent the mean ± SD of 3 independent experiments, *, P < 0.05, **, P < 0.01. (C)

QPCR was performed to determine gene levels in mouse CD138+ cells, with GAPDH

used as an internal normalization control. Error bars represent the mean ± SD of 3

independent experiments, **: p < 0.01, ***: p < 0.001 versus vehicle control. (D)

Survival curves (Kaplan-Meier) of mice showed prolongation of overall survival with

indicated treatments (log-rank test, p < 0.01).

Figure 6. Exosomal PSMA3 and PSMA3-AS1 levels in plasma and CD138+ MM

34

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

cells correlate with BTZ response in MM Patients. (A) (B) Characterization of

circulating exosomes from the plasma of human subjects. (A) Representative TEM

imaging of exosomes from the plasma of human subjects (Bar = 100 nm). (B)

Exosomes from the plasma of human subjects were measured by DLS. (C) The

number of exosomes isolated from bortezomib resistant patients and bortezomib

sensitive patients was determined. SP: bortezomib sensitive patients, RP: bortezomib

resistance patients. (D) Circulating PSMA3 and PSMA3-AS1 predominantly exists in

exosomes. Exosomes isolated from three cases of MM patient plasma as well as the

corresponding exosome depleted plasma were subjected to RNA extraction and

RT-qPCR. The ratio of mRNAs level in the exosomes to those in exosome-free

fraction was shown. Relative gene expression values in the same volume of plasma

were normalized to exogenous reference λ polyA. (E) Pearson correlation analysis

between PSMA3 and PSMA3-AS1 levels in CD138+ myeloma cells and that in

circulating exosomes of MM patients (n = 57). Relative gene expression values in the

same volume of plasma were normalized to exogenous reference λ polyA. Results are

presented as mean ± -AS1 on

overall progression-free survival (PFS) and survival (OS). (G) The prognostic impact

of exosomal PSMA3 on overall progression-free survival (PFS) and survival (OS). (H)

A schematic diagram depicting the mechanism of transferring of PSMA3 and

PSMA3-AS1 by exosome conferring PIs resistance to MSCs.

35

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.
Author Manuscript Published OnlineFirst on January 4, 2019; DOI: 10.1158/1078-0432.CCR-18-2363
Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Exosome-Transmitted PSMA3 and PSMA3-AS1 Promotes


Proteasome Inhibitors Resistance in Multiple Myeloma
Hongxia Xu, Huiying Han, Sha Song, et al.

Clin Cancer Res Published OnlineFirst January 4, 2019.

Updated version Access the most recent version of this article at:
doi:10.1158/1078-0432.CCR-18-2363

Supplementary Access the most recent supplemental material at:


Material http://clincancerres.aacrjournals.org/content/suppl/2019/01/04/1078-0432.CCR-18-2363.DC1

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet
Manuscript been edited.

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Subscriptions Department at pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://clincancerres.aacrjournals.org/content/early/2019/01/04/1078-0432.CCR-18-2363.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
Rightslink site.

Downloaded from clincancerres.aacrjournals.org on January 6, 2019. © 2019 American Association for Cancer
Research.

You might also like