Open Access

Identification and expression of MMSA-8, and its clinical significance in multiple myeloma

  • Authors:
    • Rui He
    • Nan Yang
    • Pengyu Zhang
    • Jie Liu
    • Junhui Li
    • Fulin Zhou
    • Wanggang Zhang
  • View Affiliations

  • Published online on: April 28, 2017     https://doi.org/10.3892/or.2017.5609
  • Pages: 3235-3243
  • Copyright: © He et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In our previous studies, we identified 12 multiple myeloma (MM)-associated antigens by serological analysis of tumor-associated antigens with a recombinant cDNA expression library (SEREX) on MM. MM-associated antigen-8 (MMSA-8) was one of the new antigens identified. We determined the 3'- and 5'-ends of MMSA-8 using SMART-rapid amplification of cDNA ends (RACE) and then cloned its full-length cDNA in the U266 cell line. The full cDNA sequence revealed that MMSA-8 is RPS27A-related transcript variant 1 that is specifically associated with MM. We examined its prognostic significance for the first time, by investigating the correlations between MMSA-8 expression and definite clinicopathological features. We quantitatively assessed MMSA-8 expression using qRT-PCR and western blot analysis in healthy donors and MM patients. The expression levels of MMSA-8 were upregulated with statistical significance in MM patients in contrast to those in healthy donors. The expression of MMSA-8 was also upregulated in relapsed patients compared with that in the complete remission (CR) group. Contrasting MMSA-8 expression levels in different patients with definite clinicopathological features suggested an association between MMSA-8 with unfavorable clinicopathological characteristics, such as international staging system (ISS) stage III, higher lactate dehydrogenase (LDH) levels and higher C-reactive protein (CRP) levels. The expression of MMSA-8 was also increased in patients with unfavorable cytogenetic and genetic abnormalities, including the presence of t(11;14), t(4;14), t(14;16), del(17p), del(13q) and p53 deletion, which was statistically significant. The expression of MMSA-8 exhibited significant variance in the treatment responses of the CR, PR, progression and relapse groups. Univariate and multivariate analyses revealed that high MMSA-8 values were associated with poorer progression-free survival (PFS) and overall survival (OS) in MM patients independently. In conclusion, our data indicated that MMSA-8 is an independent and unfavorable prognostic risk factor in MM; MMSA-8 is also a promising diagnostic and therapeutic target in MM patients, but further validation is needed.

Introduction

Multiple myeloma (MM), characterized by a heterogeneous clonal proliferation of plasma cells, is the second-most common hematological malignancy (1). MM is responsible for 15–20% of deaths from hematological malignancy (1). Targeted therapies such as the proteasome inhibitor (PI) bortezomib (Bz), the immunomodulatory drugs (IMiDs) lenalidomide and thalidomide (Th) have vastly improved the prognosis of MM patients (24). Unfortunately, MM is a heterogeneous disease with various disease courses, responses to therapy and survival outcome, and many patients still have to face challenges from disease progression (DP) and eventually succumb to MM (5). Thus, research to identify novel therapeutic agents is urgently needed. One promising area of recent investigation is the development of immunotherapies that target and eliminate myeloma cells more selectively (611). The premise of these therapies is the identification of cancer-specific and/or cancer-associated antigens (CAA) through the immune system. Moreover, finding an effective serological molecular marker which can forecast the prognoses and therapeutic responses of MM patients will be a valuable tool for MM therapy.

MM-associated antigen-8 (MMSA-8) (GenBank no. AY952889.1) is a new MM-associated antigen (MAA) that we previously identified. We serologically assessed tumor antigens using a recombinant cDNA expression library (SEREX) of MM to identify MM-associated antigens (MMSAs). MMSA-8 was confirmed as a new antigen that specifically interacted with the serum from patients with allogeneic myeloma but did not interact with normal donors (12). Differences in the MMSA-8 sequence with the novel available working version of the human genome suggests that MMSA-8 is a new RPS27A-related transcription variant that is specifically associated with MM. The corresponding gene is human RPS27A, which is located on chromosome 2p16. RPS27A transcript variant 1, RPS27A transcript variant 2 and RPS27A transcript variant 3 are distinct transcripts of RPS27A, but they encode the same protein (13,14). Further exploration of the correlation between MMSA-8 and RPS27A and of MMSA-8 expression patterns in MM thus became necessary. Therefore, the full-length cDNA sequence of MMSA-8 was cloned and its expression pattern was investigated in MM patients.

Materials and methods

Cell line culture

U266 cells were purchased from the Institute for Cancer Research of the Medical School of Xi'an Jiaotong University (China). The cells were cultured with RPMI-1640 medium (Gibco-BRL, Life Technologies Ltd., Paisley, Scotland) that was supplemented with 15% fetal bovine serum (FBS), 0.1 mg/ml streptomycin and 100 U/ml penicillin G and incubated at 37°C in a humidified atmosphere with 5% CO2. Cells in a log phase of growth were used for all experiments.

Patients

Fifteen healthy hematopoietic stem cell transplant donors were obtained as controls. The donors and 85 MM patients were enrolled in the study at the Department of Clinical Hematology of the Second Affiliated Hospital, Medical School of Xi'an Jiaotong University, between March 2011 and December 2014. Diagnoses were based on the 2008 World Health Organization criteria. The distribution of the clinical parameters and clinicopathological features of the patients are displayed in Table I. Protocols and response definitions were based on the 2011 MM Clinical Practice Guidelines of the National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines in Oncology. Patients received drugs, including Bz, Th, dexamethasone (D), vincristine (V), adriamycin (A) and prednisolone (P), in varying combinations. Thirty five patients used VAD (V + A + D) regimens, 30 patients used PD (Bz + D) regimens, 16 patients used a Th-Bz combination and 4 patients used PAD (Bz + A + D) regimens combination as induction therapy. Th (100–200 mg) was taken orally each day during chemotherapy intervals. Most patients received a Th-P combination as maintenance therapy. Therapeutic responses were defined as complete remission (CR), partial remission (PR), progression or relapse. Follow-ups were performed on 68 MM patients, and the 17 MM patients were assessed at the first time of diagnosis and relapse. The duration of the median follow-up was 25.5 months, with a range of 5 to 45 months. All collected samples had the written informed consent of the subjects. The Ethics Committee of the School of Medicine of Xi'an Jiaotong University approved the study and all procedures were conducted in compliance with the Declaration of Helsinki.

Table I.

The expression rates and expression level of MMSA-8 with respect to the clinical characteristics of the MM cases.

Table I.

The expression rates and expression level of MMSA-8 with respect to the clinical characteristics of the MM cases.

Expression rates of MMSA-8

CharacteristicsTotal, N=85Positive n=60Negative n=25P-valueExpression level of MMSA-8P-value
Sex, female/male37/4828/329/16   0.47340.05/50.680.601
Age (years), ≥65/<6520/6514/466/9   1.00054.60/43.420.569
ISS stage
  I23  914   0.008   3.26<0.001
  II392910   0.04239.90<0.001
  III2322  1<0.00199.28<0.001
Isotype   0.578
  igG423111 48.400.899
  igA2618  8 47.54
  igM11  7  4 38.46
  igE  1  1  0 50.10
  Light chain  5  3  2 34.52
Bone lesions
  02416  8   0.38344.180.294
  1–33628  8   0.26158.130.092
  >32516  9   1.00030.450.412
Renal failure
  (<2.0/≥2.0 Scr mg/dl)74/1150/424/7   0.08948.24/31.320.323
Hb (>85/≤85 g/l)68/1744/1624/1   0.01844.84/50.910.229
Calcium (mg/dl) (<12/≥12)69/1646/1425/2   0.13347.04/41.790.609
CRP (≤10/>10 mg/l)41/4424/3617/8   0.03117.83/72.35<0.001
LDH (<250/≥250 IU/l)44/4123/3718/4<0.00115.43/78.91<0.001
Cytogenetics
  Absent281018   8.63
  Hyperdiploid  6  4  20.202 (vs. absent)   24.460.231
  t(11;14)  8  7  10.016 (vs. absent)   44.990.013
  t(4;14)  3  3  00.064 (vs. absent)   60.560.035
  t(14;16)  3  3  00.064 (vs. absent)   56.110.027
  del (17p)10  9  10.008 (vs. absent)   72.81<0.001
  del (13q)1515  0<0.001(vs. absent)   93.06<0.001
  Double abnormalities  6  6  00.006 (vs. absent)104.00<0.001
  Not examined  6
p53 deletion
  Absent714922   0.09840.600.001
  Present  8  80 120.44
  Not examined  6
Myeloma cells (<10/≥10%)34/5118/4216/9   0.0078.35/71.19<0.001

[i] MMSA-8, multiple myeloma-associated antigen-8; ISS, international staging system; CRP, C-reactive protein; LDH, lactate dehydrogenase.

Characterization of the MMSA-8 transcript using 3′- and 5′-RACE

To characterize the intact MMSA-8 transcript, 3′- and 5′- rapid amplification of cDNA ends (RACE) was performed using the SMARTer™ RACE cDNA Amplification kit (Clontech Laboratories, Inc., Mountain View, CA, USA) according to the manufacturer's instructions. The existing MMSA-8 sequences were used as a guide to design primers using Primer Premier 5.0 (12). Briefly, total RNA was isolated using TRIzol (Gibco-BRL, NY, USA). Total RNA (1 µg) was used to generate 3′- or 5′-RACE-ready cDNA using special primers, and associated reagents provided in the Moloney Murine Leukemia Virus (MMLV) reverse transcriptase kit. The nested PCR reactions for 5′- and 3′-RACE began with primary PCR, consisting of incubation at 95°C for 3 min before 33 cycles of denaturation at 94°C for 30 sec, annealing at 58°C for 30 sec, an extension at 72°C for 60 sec and a reconditioning step at 72°C for 7 min. Primary PCR was followed by secondary PCR, consisting of the same process as the primary PCR. The 3′- and 5′-RACE products were electrophoretically separated in a 1% agarose gel with 1X TAE buffer. The resulting bands were eluted with the Agarose Gel DNA Purification kit (Takara, Dalian, China) and were then cloned into the pGEM-T Easy vector (Promega, MI, USA). Four clone products from the same band were then sequenced. The DNA sequencing service was executed at Sangon Biological Engineering Technology and Service Co., Ltd. (Shanghai, China). Its corresponding primer pairs are listed in Table II.

Table II.

Primers used in the study.

Table II.

Primers used in the study.

PrimerTechniqueSequence 5′-3′
RT primer TCTTGTGCTTATTCTTCTTGGGAGTGGTG
GSP15′-RACE TTCCTTATCCTGGATCTTGGCCTTTACAT
GSP25′-RACE AGAAAGAAGGTTAAGCTGGCTGTCCTGAA
GSP33′-RACE CTTGGCCTTTACATTTTCTATCGTATCCG
GSP43′-RACE GTGCTGGGGTGTTTATGGCAAGTCA
MMSA-8FDNA-cloning ATGTGGTGCTGGGGTGTTTAT
MMSA-8RDNA-cloning TTACTTGTCTTCTGGTTGGTGAAC
β-actinFqPCR TAGTTGCGTTACACCCTTTCTTG
β-actinRqPCR TCACCTTCACCGTTCCAGTTT
MMSA-8FqPCR ACCCTCGGATACGATAGAAAATG
MMSA-8RqPCR CACCACCACGAAGTCTCAACA

[i] MMSA-8, multiple myeloma-associated antigen-8. RACE, rapid amplification of cDNA ends.

Isolation and sequencing of MMSA-8 transcript

We used the sequences of the 3′- and 5′-RACE products of MMSA-8 as a guide for designing primers to isolate the full length sequence of MMSA-8. RT-PCR was performed with the High Fidelity Prime Script™ RT-PCR kit (Takara). The product was electrophoretically separated in a 1% agarose gel with 1X TAE buffer. The resulting bands were cloned and the four clone products from each of the bands were then sequenced as previously described. Its corresponding primer pairs are listed in Table II.

Bioinformatics

Our newly cloned full-length sequences were then analyzed using Basic Local Alignment Search Tool (BLAST) search with GenBank at the NCBI. The potential coding sequences were validated with the open reading frame (ORF) finder of the NCBI.

Real-time quantitative PCR assay

Leukocyte cells were isolated from bone marrow samples using density gradient centrifugation with Ficoll-Hypaque. Total RNA was extracted from 1×106 leukocytes using TRIzol (Gibco-BRL, Gaithersburg, MD, USA). RNA quality was visually assessed by confirmation of intact 28 and 18S ribosomal bands following agarose gel electrophoresis and ethidium bromide staining. cDNA was synthesized using the RevertAid First Strand cDNA synthase kit (Fermentas, Lithuania) according to the manufacturer's instructions. Primers for qRT-PCR were designed using Primer Premier 5.0 software. Human β-actin primers were used as an internal control. qRT-PCR was performed using the SYBR-Green PCR kit (Takara, Japan) according to the manufacturer's instructions on an ABI PRISM 7500 real-time PCR system. The following thermal cycling conditions were used: 95°C for 3 min, followed by 40 cycles of 7 sec at 95°C and 10 sec at 57°C. The qRT-PCR reactions were performed in total volumes of 20 µl that contained 2 µl of sample cDNA and 0.4 µM of each primer. The resulting qRT-PCR products were 151 and 176 bp, respectively. The comparative CT method was used, and the data are presented as 2−ΔΔCT (15). Its corresponding primer pairs are listed in Table II.

Western blotting assay

Total protein was isolated and extracted with RIPA buffer (Pierce, Rockford, IL, USA) for 15 min on ice. Protein concentrations were detected using the BCA assay (Pierce). Blotted with specific anti-MMSA-8 primary Ab [anti-RPS27A antibody ab111598 (Abcam, Cambridge, MA, USA)]. Incubated in secondary antibody and then detected with enhanced chemiluminescence technique (Amersham Pharmacia Biotech). The protein values of MMSA-8 and β-actin were quantified by Quantity One 4.2.2 Software (Bio-Rad, Hercules, CA, USA).

Statistical analysis

Mann-Whitney U tests and Fisher's exact test were used. Spearman correlation was used to detect the relationship between the expression value and levels of MMSA-8 and the clinical parameters. The Kaplan-Meier method was used to calculate the progression-free survival (PFS) and overall survival (OS). Differences between survival curves were analyzed using the log-rank test. With the Cox proportional hazards model, a multivariate analysis of survival time was performed to identify the adjusted impact of MMSA-8 overexpression on PFS and OS. The SPSS software package (SPSS17.0) was used for evaluation of statistical analyses and P<0.05 was considered as statistically significant.

Results

Sequence analysis

The MMSA-8 sequence from the cDNA libraries may not represent full-length and/or mature transcripts. Therefore, we detected the 3′- and 5′-ends of MMSA-8 using SMART-RACE with the U266 cell line. We then performed RT-PCR to clone and confirm the new, full-length MMSA-8 cDNA sequence by primers that were designed using the newly obtained 3′- and 5′-RACE sequences from the U266 cell line. We did not detect other variants of MMSA-8 in the U266 cells. Therefore, we identified a full-length MMSA-8 mRNA of 1,063 bp in the U266 cells via assembly of all the RACE and RT-PCR clones. We analyzed our sequence using BLAST search on GenBank of the NCBI. The corresponding mRNA was assigned as Homo sapien ribosomal protein S27a (RPS27A), transcript variant 1 (NCBI reference sequence: NM_002954.5). The cDNA sequence of MMSA-8 was 100% identical to RPS27A, transcript variant 1. The corresponding gene was assigned as RPS27A [Homo sapien (human)] and gene ID: 6233. These results confirmed that MMSA-8 is RPS27A-related transcript variant 1.

MMSA-8 expression in MM patients

MMSA-8 mRNA and protein levels were assessed in healthy donors and in newly diagnosed, relapsed and CR groups using qRT-PCR and western blotting. The mean relative value of MMSA-8 expressed in the control group was 9.10E-01. An MMSA-8 expression value that was one log grade higher than the expression value in the control group was defined as positive; lower values were considered to be negative. MMSA-8 transcript levels revealed a striking increase in patients with DP (i.e., newly diagnosed or relapsed) relative to normal donors (P<0.001 for both groups; Fig. 1A). Greater upregulation of MMSA-8 mRNA expression was observed in relapsed patients relative to the CR group (P<0.001, Fig. 1A). There was also a significant statistical difference between healthy donors and the CR group (P=0.03; Fig. 1A). Western blot assay detected MMSA-8 protein in most MM patient samples (Fig. 2). β-actin expression was used as a loading control. A significant correlation was detected between MMSA-8 protein levels and MMSA-8 mRNA expression levels (r=0.813, P<0.001) (data not shown).

Association of MMSA-8 expression levels with patient clinicopathological features

Table I presents the clinicopathological characteristics of the MM patients at diagnosis. The expression values of MMSA-8 were significantly correlated with international staging system (ISS) stage, myeloma cell burden, lactate dehydrogenase (LDH) and C-reactive protein (CRP) levels (Table I). The expression levels of MMSA-8 were higher in patients with unfavorable cytogenetic abnormalities, including the presence of t(11;14), t(4;14), t(14;16), del(17p), del(13q) and double abnormalities; this correlation was statistically significant (Table I). There were no significant relationships between mRNA expression of MMSA-8 and age, sex, immunoglobulin isotype, bone lesions, renal failure, anemia, hypercalcemia or hyperdiploid abnormalities (Table I).

Association of MMSA-8 expression with therapeutic response

The association of MMSA-8 expression with treatment response was examined; treatment responses were classified as CR, PR, progression or relapse. The mean MMSA-8 levels were 5.11E+01 in patients with PR, 6.58E+01 in patients with DP and 9.93E+01 in patients with relapse. The three groups exhibited much higher MMSA-8 expression than the CR group (P=0.001, P<0.001 and P<0.001, respectively; Fig. 1B). A significant difference between the PR and relapse groups was also found (P=0.004; Fig. 1B).

Association of MMSA-8 expression with p53 deletion

Table I shows the prevalence of the p53 deletion in MM patients, as assessed using fluorescence in situ hybridization (FISH) at diagnosis. The p53 deletion was detected in 8 of the 79 patients, and all 8 (100%) of the p53-deletion patients were positive for MMSA-8 expression. The positive MMSA-8 expression rates were lower in patients without p53 deletion (69.01%), although this tendency was not statistically significant (P=0.098). The level of MMSA-8 expression was higher in groups with the p53 deletion than in groups lacking this genetic aberration; this tendency was statistically significant (P=0.001; Table I).

Correlation of MMSA-8 expression with clinical outcome

The median duration of follow-up was 25.5 months, with a range of 5 to 45 months. MM relapsed and progressed in 29 patients, and therapeutic mortality was observed in 25 patients. The rates of OS and PFS were much lower in the MMSA-8-positive group (25% OS and 0% PFS) than in the MMSA-8-negative group (60% OS and 11.4% PFS, P<0.001 for both; Fig. 3). Patients in the MMSA-8-positive group had a significantly shorter PFS (median 11.72 months) than patients in the MMSA-8-negative group (median 28.53 months, P<0.001; Fig. 3A). The OS was significantly shorter (median 27.05 months) for patients in the MMSA-8-positive group than for patients in the MMSA-8-negative group (median 41.23 months, P<0.001; Fig. 3B). With univariate risk factor analysis, including age, sex, ISS stage, immunoglobulin isotype, bone lesions, renal failure, Hb, hypercalcemia, CRP, LDH, p53 deletion, myeloma cell burden, t(4;14), t(11;14), t(14;16), del(17p), del(13q), double cytogenetic abnormalities and MMSA-8 expression level it was revealed that ISS stage, CRP, LDH, myeloma cell burden, del(17P), del(13q), double cytogenetic abnormalities, p53 deletion and MMSA-8 expression were significant (Table III). A multivariate analysis confirmed myeloma cell burden, p53 deletion and MMSA-8 expression level as risk factors for PFS and OS (Table IV).

Table III.

Univariate analysis of PFS and OS in MM patients.

Table III.

Univariate analysis of PFS and OS in MM patients.

P-valuea

VariablePFSOS
Sex
  Female/male   0.735   0.957
Age (years)
  ≥65 vs. <65   0.975   0.213
ISS stage
  I vs. II and III<0.001<0.001
Immunoglobulin isotype
  IgG/IgA/IgM/IgE/light chain only   0.665   0.905
Bone lesions
  0/1–3/>3   0.579   0.584
Renal failure (Scr mg/dl)
  <2.0 vs. ≥2.0   0.742   0.745
Hb (g/l)
>85 vs. ≤85   0.499   0.253
Hypercalcemia calcium (mg/dl)
  <12 vs. ≥12   0.494   0.864
CRP (mg/l)
  ≤10 vs. >10   0.004   0.012
LDH (IU/L)
  <250 vs. ≥250<0.001   0.002
Myeloma cell burden (%)<0.001<0.001
t(11;14)
  Absent vs. present   0.909   0.908
t(14;16)
  Absent vs. present   0.789   0.857
t(4;14)
  Absent vs. present   0.568   0.917
Hyperdiploid
  Absent vs. present   0.328   0.393
Del(17p)
  Absent vs. present   0.051   0.040
Del(13q)
  Absent vs. present<0.001   0.028
Double abnormalities
  Absent vs. present   0.028   0.055
p53 deletion
  Absent vs. present<0.001   0.001
MMSA-8 expression level
  Negative vs. positive<0.001<0.001

a Log-rank test. MM, multiple myeloma; PFS, progression-free survival; OS, overall survival; ISS, international staging system; CRP, C-reactive protein; LDH, lactate dehydrogenase; MMSA-8, multiple myeloma-associated antigen-8.

Table IV.

Multivariate analysis of PFS and OS in MM patients.

Table IV.

Multivariate analysis of PFS and OS in MM patients.

PFSOS


VariablesP-valueRR (95% CI)P-valueRR (95% CI)
ISS stage0.051 0.054
  I vs. II and III
  CRP (mg/l)
  ≤10 vs. >100.3541.385 (0.696–2.758)0.2740.479 (0.128–1.792)
LDH (IU/l)
  <250 vs. ≥2500.7851.109 (0.529–2.323)0.3570.546 (0.151–1.978)
Myeloma cell burden (%)0.0145.908 (1.437–24.291)0.00731.102 (2.598–372.374)
Del(17p)
  Absent vs. present0.1071.919 (0.869–4.241)0.2002.288 (0.645–8.113)
Del(13q)
  Absent vs. present0.9651.018 (0.452–2.294)0.1150.275 (0.055–1.372)
Double abnormalities
  Absent vs. present0.7291.190 (0.445–3.180)0.0565.761 (0.955–34.747)
P53 deletion
  Absent vs. present0.0083.775 (1.410–10.102)0.00112.851 (2.948–56.017)
MMSA-8 expression level
  Negative vs. positive<0.0017.093 (2.887–17.427)0.0338.406 (1.191–59.314)

[i] PFS, progression-free survival; OS, overall survival; MM, multiple myeloma; ISS, international staging system; CRP, C-reactive protein; LDH, lactate dehydrogenase; MMSA-8, multiple myeloma-associated antigen-8; RR, risk ratio.

Discussion

MMSA-8 is an MM-associated antigen that was identified by SEREX. The present study first determined the 3′- and 5′-ends of the MMSA-8 cDNA sequence in MM using SMART-RACE. The MMSA-8 cDNA sequence was fully concordant with RPS27A transcript variant 1(13,14). The RPS27A gene encodes a fusion protein comprised of ubiquitin (Ub) at the N terminus and ribosomal protein (RP) S27a at the C terminus. RPS27a is a ribosomal protein with a Ub C-terminal extension. The Ub-RPS27a precursor protein is rapidly processed by hydrolysis into an individual Ub monomer (1618). RPS27a may perform extra-ribosomal functions in addition to its role in ribosome biogenesis and in post-translational protein modification. RPS27a is overexpressed in mouse liver cancer and in some human tumors (1921). The exact extra-ribosomal function of RPS27a is not clear. Expression of RPS27a was increased in HMy2 cells, PRMI8226 cells and U266 cells in the present study. RNA interference (RNAi) technology demonstrated that MMSA-8 downregulation inhibited U266 cell growth, increased cell apoptosis and the number of cells in the S phase. Our results suggest that MMSA-8 exerts a significant role in the generation and progression of MM.

This study investigated MMSA-8 mRNA and protein expression in MM patients and normal donors, and its relationship with certain clinicopathological characteristics in prognosis. We detected that the expression levels of MMSA-8 were significantly upregulated in MM patients compared with those in normal donors and that relapsed patients exhibited a greater upregulation of MMSA-8 expression compared to the CR group. These results confirmed our previous hypothesis that MMSA-8 expression is closely related to DP. The difference in the expression of MMSA-8 in various characteristic groups indicated its unfavorable associated clinicopathological characteristics. Age, ISS stage, bone lesions and creatinine, LDH, CRP, Hb and calcium levels are independent prognostic factors of MM, and the present study indicated that patients in ISS stage III with higher LDH or CRP levels also had higher MMSA-8 expression levels (2225). The MMSA-8 value was also higher in patients with unfavorable cytogenetic abnormalities, including the presence of t(11;14), t(4;14), t(14;16), del(17p), del(13q) and double abnormalities, and this association was statistically significant (2630). However, there was no statistically significant association between MMSA-8 expression and age, anemia or hypercalcemia, thereby contradicting some previously published studies (31,32). This discrepancy may be the result of the multiplicity and heterogeneity of myeloma cells in our studies (33). Positive MMSA-8 expression was associated with unfavorable clinical features at diagnosis. Strong evidence confirms that MMSA-8 is an unfavorable prognostic factor in MM. We also observed the lowest MMSA-8 mRNA expression level in the CR group, compared to patients with PR and DP. Relapsed patients exhibited the highest MMSA-8 expression, further supporting the hypothesis that MMSA-8 is a strong prognostic factor of response to therapy in MM patients.

Deletion of p53 was detected in 8 of the 79 patients in our study and was associated with increased MMSA-8 mRNA levels, suggesting that MMSA-8 is related to p53 deletion. p53 is a tumor-suppressor gene that has been involved in the control of cell proliferation, differentiation, invasion and apoptosis (34). p53 gene deletions in MM are associated with poor patient survival (28,3537). As a direct transcriptional target of p53, the RPS27A gene is often overexpressed in response to DNA damage (38). RPS27a also interacts with MDM2 to suppress MDM2-associated p53 ubiquitination and then leading to the activation of p53 and cell cycle arrest (39). RPS27a may perform extra-ribosomal functions in addition to its role in ribosome biogenesis and post-translational protein modification (40,41). Previously published research support our observations and conclusion, but the intrinsic mechanism involved in p53-RPS27a interactions requires further investigation (39).

We also analyzed the correlation between MMSA-8 expression and clinical outcome. The results demonstrated that the patients in the MMSA-8-positive group had lower rates of PFS and OS than the MMSA-8-negative patients. Patients with positive MMSA-8 expression also had significantly shorter PFS and OS. Univariate risk factor analysis found that ISS stage, CRP, LDH, myeloma cell burden, del(17P), del(13q), double cytogenetic abnormalities, p53 deletion and MMSA-8 expression were significantly significant. Multivariate analysis revealed that only the expression level of MMSA-8, p53 deletion and myeloma cell burden were independent risk factors in MM patients. These findings are consistent with previously published studies (4244). However, we are the first to observe the relevance of the expression level of MMSA-8 to patient survival.

In conclusion, the full-length cDNA sequence of MMSA-8 was cloned in MM and it was hypothesized that MMSA-8 is MM-associated RPS27A transcript variant 1. We firstly demonstrated the specific expression of MMSA-8 in MM patients and the association of its dysregulation with unfavorable clinical features at diagnosis and poorer therapeutic outcome in MM. Univariate and multivariate analyses revealed that MMSA-8 may be an independent prognostic factor in MM. The present study supports further investigation to ascertain MMSA-8 as a promising diagnostic marker and therapeutic target in MM. Future studies should focus on its pathophysiological relevance in myeloma cells, especially with regard to its interaction with p53.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (no. 81172257).

References

1 

Group IMW; International Myeloma Working Group, : Criteria for the classification of monoclonal gammopathies, multiple myeloma and related disorders: A report of the International Myeloma Working Group. Br J Haematol. 121:749–757. 2003. View Article : Google Scholar : PubMed/NCBI

2 

Kumar SK, Rajkumar SV, Dispenzieri A, Lacy MQ, Hayman SR, Buadi FK, Zeldenrust SR, Dingli D, Russell SJ, Lust JA, et al: Improved survival in multiple myeloma and the impact of novel therapies. Blood. 111:2516–2520. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Pulte D, Gondos A and Brenner H: Improvement in survival of older adults with multiple myeloma: Results of an updated period analysis of SEER data. Oncologist. 16:1600–1603. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Brenner H, Gondos A and Pulte D: Expected long-term survival of patients diagnosed with multiple myeloma in 2006–2010. Haematologica. 94:270–275. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Kumar SK, Lee JH, Lahuerta JJ, Morgan G, Richardson PG, Crowley J, Haessler J, Feather J, Hoering A, Moreau P, et al International Myeloma Working Group, : Risk of progression and survival in multiple myeloma relapsing after therapy with IMiDs and bortezomib: A multicenter international myeloma working group study. Leukemia. 26:149–157. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Tai YT, Li X, Tong X, Santos D, Otsuki T, Catley L, Tournilhac O, Podar K, Hideshima T, Schlossman R, et al: Human anti-CD40 antagonist antibody triggers significant antitumor activity against human multiple myeloma. Cancer Res. 65:5898–5906. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Bae J, Prabhala R, Voskertchian A, Brown A, Maguire C, Richardson P, Dranoff G, Anderson KC and Munshi NC: A multiepitope of XBP1, CD138 and CS1 peptides induces myeloma-specific cytotoxic T lymphocytes in T cells of smoldering myeloma patients. Leukemia. 29:218–229. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Rosenblatt J, Avivi I, Vasir B, Uhl L, Munshi NC, Katz T, Dey BR, Somaiya P, Mills H, Campigotto F, et al: Vaccination with dendritic cell/tumor fusions following autologous stem cell transplant induces immunologic and clinical responses in multiple myeloma patients. Clin Cancer Res. 19:3640–3648. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Hong S, Li H, Qian J, Yang J, Lu Y and Yi Q: Optimizing dendritic cell vaccine for immunotherapy in multiple myeloma: Tumour lysates are more potent tumour antigens than idiotype protein to promote anti-tumour immunity. Clin Exp Immunol. 170:167–177. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Anderson LD Jr, Cook DR, Yamamoto TN, Berger C, Maloney DG and Riddell SR: Identification of MAGE-C1 (CT-7) epitopes for T-cell therapy of multiple myeloma. Cancer Immunol Immunother. 60:985–997. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Michalek J, Ocadlikova D, Matejkova E, Foltankova V, Dudová S, Slaby O, Horvath R, Pour L and Hajek R: Individual myeloma-specific T-cell clones eliminate tumour cells and correlate with clinical outcomes in patients with multiple myeloma. Br J Haematol. 148:859–867. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Zhou FL, Zhang WG, Chen G, Zhao WH, Cao XM, Chen YX, Tian W, Liu J and Liu SH: Serological identification and bioinformatics analysis of immunogenic antigens in multiple myeloma. Cancer Immunol Immunother. 55:910–917. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Lamesch P, Li N, Milstein S, Fan C, Hao T, Szabo G, Hu Z, Venkatesan K, Bethel G, Martin P, et al: hORFeome v3.1: A resource of human open reading frames representing over 10,000 human genes. Genomics. 89:307–315. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Gonzalez-Begne M, Lu B, Han X, Hagen FK, Hand AR, Melvin JE and Yates JR: Proteomic analysis of human parotid gland exosomes by multidimensional protein identification technology (MudPIT). J Proteome Res. 8:1304–1314. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Redman KL and Rechsteiner M: Identification of the long ubiquitin extension as ribosomal protein S27a. Nature. 338:438–440. 1989. View Article : Google Scholar : PubMed/NCBI

17 

Shabek N and Ciechanover A: Degradation of ubiquitin: The fate of the cellular reaper. Cell Cycle. 9:523–530. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Komander D, Clague MJ and Urbé S: Breaking the chains: Structure and function of the deubiquitinases. Nat Rev Mol Cell Biol. 10:550–563. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Adams SM, Sharp MG, Walker RA, Brammar WJ and Varley JM: Differential expression of translation-associated genes in benign and malignant human breast tumours. Br J Cancer. 65:65–71. 1992. View Article : Google Scholar : PubMed/NCBI

20 

Wong JM, Mafune K, Yow H, Rivers EN, Ravikumar TS, Steele GD Jr and Chen LB: Ubiquitin-ribosomal protein S27a gene overexpressed in human colorectal carcinoma is an early growth response gene. Cancer Res. 53:1916–1920. 1993.PubMed/NCBI

21 

Kanayama H, Tanaka K, Aki M, Kagawa S, Miyaji H, Satoh M, Okada F, Sato S, Shimbara N and Ichihara A: Changes in expressions of proteasome and ubiquitin genes in human renal cancer cells. Cancer Res. 51:6677–6685. 1991.PubMed/NCBI

22 

Vincent Rajkumar S: Multiple myeloma: 2014 update on diagnosis, risk-stratification, and management. Am J Hematol. 89:999–1009. 2014.PubMed/NCBI

23 

Tarkun P, Atalay F, Atesoglu EB, Mehtap O, Simsek M, Terzi E, Geduk A, Balli F, Batman A, Baydemir C, et al: Treatment of patients with multiple myeloma over 65 yr: More tolerability or better response? Eur J Haematol. 94:424–430. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Chim CS, Sim J, Tam S, Tse E, Lie AK and Kwong YL: LDH is an adverse prognostic factor independent of ISS in transplant-eligible myeloma patients receiving bortezomib-based induction regimens. Eur J Haematol. 94:330–335. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Kiba T, Ito T, Nakashima T, Okikawa Y, Kido M, Kimura A, Kameda K, Miyamae F, Tanaka S, Atsumi M, et al: Bortezomib and dexamethasone for multiple myeloma: Higher AST and LDH levels associated with a worse prognosis on overall survival. BMC Cancer. 14:4622014. View Article : Google Scholar : PubMed/NCBI

26 

Tricot G, Barlogie B, Jagannath S, Bracy D, Mattox S, Vesole DH, Naucke S and Sawyer JR: Poor prognosis in multiple myeloma is associated only with partial or complete deletions of chromosome 13 or abnormalities involving 11q and not with other karyotype abnormalities. Blood. 86:4250–4256. 1995.PubMed/NCBI

27 

Fonseca R, Barlogie B, Bataille R, Bastard C, Bergsagel PL, Chesi M, Davies FE, Drach J, Greipp PR, Kirsch IR, et al: Genetics and cytogenetics of multiple myeloma: A workshop report. Cancer Res. 64:1546–1558. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Fonseca R, Blood E, Rue M, Harrington D, Oken MM, Kyle RA, Dewald GW, van Ness B, van Wier SA, Henderson KJ, et al: Clinical and biologic implications of recurrent genomic aberrations in myeloma. Blood. 101:4569–4575. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Facon T, Avet-Loiseau H, Guillerm G, Moreau P, Geneviève F, Zandecki M, Laï JL, Leleu X, Jouet JP, Bauters F, et al: Intergroupe Francophone du Myélome: Chromosome 13 abnormalities identified by FISH analysis and serum beta2-microglobulin produce a powerful myeloma staging system for patients receiving high-dose therapy. Blood. 97:1566–1571. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Oh S, Koo DH, Kwon MJ, Kim K, Suh C, Min CK, Yoon SS, Shin HJ, Jo DY, Kwak JY, et al: Korean Multiple Myeloma Working Party (KMMWP): Chromosome 13 deletion and hypodiploidy on conventional cytogenetics are robust prognostic factors in Korean multiple myeloma patients: Web-based multicenter registry study. Ann Hematol. 93:1353–1361. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Kyrtsonis MC, Maltezas D, Tzenou T, Koulieris E and Bradwell AR: Staging systems and prognostic factors as a guide to therapeutic decisions in multiple myeloma. Semin Hematol. 46:110–117. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Younes M, Hachfi H, Hammouda F, Younes K, Ben Hammouda S, Jguirim M, Zrour S, Béjia I, Touzi M and Bergaoui N: Survival prognosis factors in multiple myeloma. Tunis Med. 92:399–405. 2014.(In French). PubMed/NCBI

33 

Lohr JG, Stojanov P, Carter SL, Cruz-Gordillo P, Lawrence MS, Auclair D, Sougnez C, Knoechel B, Gould J, Saksena G, et al: Multiple Myeloma Research Consortium: Widespread genetic heterogeneity in multiple myeloma: Implications for targeted therapy. Cancer Cell. 25:91–101. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Levine AJ: p53, the cellular gatekeeper for growth and division. Cell. 88:323–331. 1997. View Article : Google Scholar : PubMed/NCBI

35 

Drach J, Ackermann J, Fritz E, Krömer E, Schuster R, Gisslinger H, DeSantis M, Zojer N, Fiegl M, Roka S, et al: Presence of a p53 gene deletion in patients with multiple myeloma predicts for short survival after conventional-dose chemotherapy. Blood. 92:802–809. 1998.PubMed/NCBI

36 

Königsberg R, Zojer N, Ackermann J, Krömer E, Kittler H, Fritz E, Kaufmann H, Nösslinger T, Riedl L, Gisslinger H, et al: Predictive role of interphase cytogenetics for survival of patients with multiple myeloma. J Clin Oncol. 18:804–812. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Chang H, Qi C, Yi QL, Reece D and Stewart AK: p53 gene deletion detected by fluorescence in situ hybridization is an adverse prognostic factor for patients with multiple myeloma following autologous stem cell transplantation. Blood. 105:358–360. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Nosrati N, Kapoor NR and Kumar V: DNA damage stress induces the expression of ribosomal protein S27a gene in a p53-dependent manner. Gene. 559:44–51. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Sun XX, DeVine T, Challagundla KB and Dai MS: Interplay between ribosomal protein S27a and MDM2 protein in p53 activation in response to ribosomal stress. J Biol Chem. 286:22730–22741. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Warner JR and McIntosh KB: How common are extraribosomal functions of ribosomal proteins? Mol Cell. 34:3–11. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Wool IG: Extraribosomal functions of ribosomal proteins. Trends Biochem Sci. 21:164–165. 1996. View Article : Google Scholar : PubMed/NCBI

42 

Gertz MA, Lacy MQ, Dispenzieri A, Greipp PR, Litzow MR, Henderson KJ, van Wier SA, Ahmann GJ and Fonseca R: Clinical implications of t(11;14)(q13;q32), t(4;14)(p16.3;q32), and −17p13 in myeloma patients treated with high-dose therapy. Blood. 106:2837–2840. 2005. View Article : Google Scholar : PubMed/NCBI

43 

van de Donk NW and Sonneveld P: Diagnosis and risk stratification in multiple myeloma. Hematol Oncol Clin North Am. 28:791–813. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Mangiacavalli S, Pochintesta L, Cocito F, Pompa A, Bernasconi P, Cazzola M and Corso A: Correlation between burden of 17P13.1 alteration and rapid escape to plasma cell leukaemia in multiple myeloma. Br J Haematol. 162:555–558. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2017
Volume 37 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He R, Yang N, Zhang P, Liu J, Li J, Zhou F and Zhang W: Identification and expression of MMSA-8, and its clinical significance in multiple myeloma. Oncol Rep 37: 3235-3243, 2017
APA
He, R., Yang, N., Zhang, P., Liu, J., Li, J., Zhou, F., & Zhang, W. (2017). Identification and expression of MMSA-8, and its clinical significance in multiple myeloma. Oncology Reports, 37, 3235-3243. https://doi.org/10.3892/or.2017.5609
MLA
He, R., Yang, N., Zhang, P., Liu, J., Li, J., Zhou, F., Zhang, W."Identification and expression of MMSA-8, and its clinical significance in multiple myeloma". Oncology Reports 37.6 (2017): 3235-3243.
Chicago
He, R., Yang, N., Zhang, P., Liu, J., Li, J., Zhou, F., Zhang, W."Identification and expression of MMSA-8, and its clinical significance in multiple myeloma". Oncology Reports 37, no. 6 (2017): 3235-3243. https://doi.org/10.3892/or.2017.5609