Background: Aberrantly expressed miRNAs promote renal cell carcinoma (RCC) growth and metastasis and are potentially useful biomarkers for metastatic disease. However, a consensus clinically significant miRNA signature has not been identified. To identify an miRNA signature for predicting clinical outcome in RCC patients, we used a four-pronged interconnected approach.

Methods: Differentially expressed miRNAs were identified and analyzed in 113 specimens (normal kidney: 59; tumor: 54). miRNA profiling was performed in matched normal and tumor specimens from 8 patients and extended to 32 specimens. Seven aberrantly expressed miRNAs were analyzed by qPCR, and their levels were correlated with RCC subtypes and clinical outcome. miRNA signature was confirmed in The Cancer Genome Atlas RCC dataset (n = 241).

Results: Discovery phase identified miR-21, miR-142-3p, miR-142-5p, miR-150, and miR-155 as significantly upregulated (2–4-fold) and miR-192 and miR-194 as downregulated (3–60-fold) in RCC; miR-155 distinguished small tumors (<4 cm) from benign oncocytomas. In univariate and multivariate analyses, miRNA combinations (miR-21+194; miR-21+142-5p+194) significantly predicted metastasis and/or disease-specific mortality; miR-21+142-5p+194 (for metastasis): P = 0.0017; OR, 0.53; 95% confidence interval (CI), 0.75–0.33; 86.7% sensitivity; 82% specificity. In the TCGA dataset, combined biomarkers associated with metastasis and overall survival (miR-21+142-5p+194: P < 0.0001; OR, 0.37; 95% CI, 0.58–0.23).

Conclusions: The interconnected discovery–validation approach identified a three-miRNA signature as a potential predictor of disease outcome in RCC patients.

Impact: With 10% survival at 5 years, metastatic disease presents poor prognosis for RCC patients. The three-miRNA signature discovered and validated may potentially at an early stage detect and predict metastasis, to allow early intervention for improving patient prognosis. Cancer Epidemiol Biomarkers Prev; 27(4); 464–72. ©2018 AACR.

Annually, about 300,000 new cases and approximately 134,000 deaths are attributed to kidney cancer worldwide (1, 2). More than 90% of kidney tumors are renal cell carcinomas (RCCs), which arise from the epithelial lining of the proximal convoluted tubule (3). RCC includes a spectrum of tumors ranging from those with indolent clinical course to those with high metastatic propensity. Clear cell RCC (ccRCC) accounts for 70% to 80% of all RCC cases and is also the most aggressive subtype, accounting for 80% to 90% of cases in metastatic disease (4), and papillary (10%–15%), chromophobe (5%–10%), benign oncocytomas, etc. account for the rest (1, 5–7). Genome-wide expression analysis further divides the histologic subtypes of RCC, such as, ccRCC, into molecular subtypes, some of which may associate with response to targeted treatments (8–10). Computerized tomography and magnetic resonance imaging allow the detection of small renal tumors (<4 cm); however, these techniques do not efficiently differentiate benign oncocytomas from malignant RCC subtypes (11–14). At initial presentation, up to one-third of RCC patients have synchronous metastasis, and up to 30% of patients develop metachronous metastasis after nephrectomy and require salvage therapy (15). The 5-year survival of patients with metastatic RCC is as low as 10%. Genomic, proteomic, and metabolomic characterization of normal and RCC tissues as well as between benign and malignant RCC may accurately predict metastasis and clinical outcome and aid in designing targeted therapies (5–7, 10, 16–18).

MiRNAs are small noncoding RNAs (19–22 nucleotides) that posttranslationally regulate gene expression by causing mRNA degradation or inhibiting their translation through RNA interference (19–22). miRNAs bind to target sequences in the 3′-untranslated region of mRNAs and cause mRNA degradation through a well-defined process. Consequence sequences recognized by specific miRNAs are shared among a few or hundreds of mRNAs. Therefore, aberrant expression of miRNAs in disease processes globally affects a variety of cellular functions and signaling pathways. In almost all malignancies, the expression pattern of miRNAs is altered. Many miRNAs are causally linked to the suppression or malignant progression of cancer, and to treatment response, due to the silencing of tumor suppressors or the upregulation of oncogenic signaling. Functional studies have confirmed the role of several miRNAs in promoting RCC growth and metastasis (23).

Using quantitative real-time PCR (qPCR), deep sequencing, and microarray techniques, several studies have identified miRNAs that are differentially expressed in RCC tissues and established their association with clinical outcome (22, 24–33). However, different studies report different miRNAs as being differentially expressed in RCC. miRNAs such as miR-141, miR-200c (miR-200 family), and miR-429 have been reported as most downregulated, whereas other studies report that decreased expression of miR-106b, miR-99a, miR-1826, miR-215, miR-217, miR-187, etc. correlates with poor prognosis (20, 21). A similar case can be made regarding the upregulation of miRNAs. Among some studies, upregulation of miR-21 and downregulation of miR-126 have been suggested as potential prognostic markers for poor outcome; however, other studies did not find this signature (19, 20, 26, 34–37). A plausible reason for different studies reporting different “clinically significant miRNAs” could be that many studies do not use matched normal and tumor specimens in the discovery phase such as miRNA profiling, which may cause non–RCC-related factors, such as comorbid conditions and genetic makeup of the subjects, to influence the results.

To identify miRNAs that might serve as potentially accurate miRNA signatures for molecular subtypes in RCC, and for predicting clinical outcome, we used a four-pronged interconnected approach. We initially performed miRNA profiling in matched normal and tumor specimens from the same patients, and then confirmed the profile by miRNA profiling of unmatched specimens. miRNAs which were significantly aberrantly expressed were validated by qPCR assays. Last, the identified and confirmed miRNA signature was further validated in The Cancer Genome Atlas (TCGA) dataset.

Tissue specimens

In clinical specimen cohort, normal kidney (n = 59) and tumor (n = 54) specimens were collected from patients undergoing radical or partial nephrectomy (laparoscopic or open) for a renal mass between July 2007 and November 2009. Among these, normal kidney and tumor specimens were collected from the same 38 patients (matched specimens). The remaining normal (n = 21) and tumor (n = 16) specimens were collected from 37 other patients (unmatched specimens). All specimens were obtained based on their availability for research purpose and under a protocol approved by University of Miami's Institutional Review Board; a written consent was obtained from study individuals. These specimens were transferred to Augusta University under an approved protocol. TCGA dataset contains 311 specimens on whom miRNA data were available, along with demographic and pathologic parameters and clinical outcome in terms of metastasis and/or survival. Among these, 70 are normal kidney specimens. The TCGA dataset did not contain information regarding time to metastasis, and data were missing for recurrence-free survival on 201 patients. Patient and tissue characteristics for both sets are presented in Table 1.

Table 1.

Patient characteristics

ParameterClinical specimen cohortTCGA dataset
Number of specimens Normal kidney: 59 Normal: 70; tumor (RCC): 241 
 Tumor: 54  
 RCC: 49 (90.7%); oncocytoma: 5 (9.3%)  
Age (y) 63.71 ± 14.86; median: 65 59.2 ± 12.2; median: 60 
Gender Male: 41 (75.9%); female: 12 (22.2%); unknown: 1 (1.9%) Tumor: Male: 160 (66.4%) female: 81 (33.6%) 
Tumor type Clear cell: 32 (59.3%); chromophobe: 4 (7.4%); papillary: 7 (type I: 5; undefined: 2; 12.9%); sarcomatoid: 6 (11.1%); oncocytoma: 5 (9.3%) Clear cell: 311 
Tumor size <4 cm: 14 (25.9%); ≥4 cm: 39 (72.2%)  
 Unknown: 1 (1.9%)  
Grade 0 (oncocytoma): 5 (9.3%) Grade: 1: 8 (3.3%); grade 2: 93 (38.6%); grade 3: 98 (40.7%); grade 4: 40 (16.6%); missing: 2 (0.8%) 
 Grade 1: 4 (7.4%); grade 2: 15 (27.8%)  
 Grade 3: 16 (29.6%); grade 4: 14 (25.9%)  
Stage pT0: 6 (11.1%); pT1a: 10 (18.5%) pT0: 3 (1.2%); pT1: 123 (51%) 
 pT1b: 8 (14.8%); pT2: 8 (14.8%) pT2: 23 (9.5%); pT3: 80 (33.3%) 
 pT3a: 11 (20.4%); pT3b: 10 (18.5%); pT4: 1 (1.9%) pT4: 12 (5%) 
Lymph node invasion (−) 28 (51.8%); (+): 7 (13%); (−): 97 (40.3%); (+): 8 (3.3%); 
 unknown: 19 (35.2%) unknown: 136 (56.4%) 
Renal vein invasion (−): 39 (72.2%); (+): 9 (16.7%);  
 Unknown: 6 (11.1%)  
Lymphovascular invasion (−) 23 (42.6%); (+): 9 (16.7%);  
 unknown: 22 (40.7%)  
Karnofsky score +: 21 (38.9%); unknown: 33 (61.1%)  
 mean: 86.67 ± 12.78; median: 90  
Metastasis (−): 39 (72.2%); (+): 15 (27.8%) (−): 169 (70.1%); (+): 42 (17.4%); unknown: 30 (12.5%) 
Follow-up (metastasis) 30.2 ± 24; 23 months  
Death (−): 15 (27.8%); (+): 8 (14.8%); missing: 31 (57.4%)  
Overall survival  (+): 167 (69.3%); (−): 72 (29.9%); Unknown: 2 (0.8%) 
Follow-up (overall survival)  45.99 ± 35.86; 38.97 months 
Recurrence-free survival  (+): 73 (30.3%); (−): 15 (6.2%); Unknown: 153 (63.5%) 
Follow-up (recurrence-free survival):  47.9± 44.3; 27.9 months 
ParameterClinical specimen cohortTCGA dataset
Number of specimens Normal kidney: 59 Normal: 70; tumor (RCC): 241 
 Tumor: 54  
 RCC: 49 (90.7%); oncocytoma: 5 (9.3%)  
Age (y) 63.71 ± 14.86; median: 65 59.2 ± 12.2; median: 60 
Gender Male: 41 (75.9%); female: 12 (22.2%); unknown: 1 (1.9%) Tumor: Male: 160 (66.4%) female: 81 (33.6%) 
Tumor type Clear cell: 32 (59.3%); chromophobe: 4 (7.4%); papillary: 7 (type I: 5; undefined: 2; 12.9%); sarcomatoid: 6 (11.1%); oncocytoma: 5 (9.3%) Clear cell: 311 
Tumor size <4 cm: 14 (25.9%); ≥4 cm: 39 (72.2%)  
 Unknown: 1 (1.9%)  
Grade 0 (oncocytoma): 5 (9.3%) Grade: 1: 8 (3.3%); grade 2: 93 (38.6%); grade 3: 98 (40.7%); grade 4: 40 (16.6%); missing: 2 (0.8%) 
 Grade 1: 4 (7.4%); grade 2: 15 (27.8%)  
 Grade 3: 16 (29.6%); grade 4: 14 (25.9%)  
Stage pT0: 6 (11.1%); pT1a: 10 (18.5%) pT0: 3 (1.2%); pT1: 123 (51%) 
 pT1b: 8 (14.8%); pT2: 8 (14.8%) pT2: 23 (9.5%); pT3: 80 (33.3%) 
 pT3a: 11 (20.4%); pT3b: 10 (18.5%); pT4: 1 (1.9%) pT4: 12 (5%) 
Lymph node invasion (−) 28 (51.8%); (+): 7 (13%); (−): 97 (40.3%); (+): 8 (3.3%); 
 unknown: 19 (35.2%) unknown: 136 (56.4%) 
Renal vein invasion (−): 39 (72.2%); (+): 9 (16.7%);  
 Unknown: 6 (11.1%)  
Lymphovascular invasion (−) 23 (42.6%); (+): 9 (16.7%);  
 unknown: 22 (40.7%)  
Karnofsky score +: 21 (38.9%); unknown: 33 (61.1%)  
 mean: 86.67 ± 12.78; median: 90  
Metastasis (−): 39 (72.2%); (+): 15 (27.8%) (−): 169 (70.1%); (+): 42 (17.4%); unknown: 30 (12.5%) 
Follow-up (metastasis) 30.2 ± 24; 23 months  
Death (−): 15 (27.8%); (+): 8 (14.8%); missing: 31 (57.4%)  
Overall survival  (+): 167 (69.3%); (−): 72 (29.9%); Unknown: 2 (0.8%) 
Follow-up (overall survival)  45.99 ± 35.86; 38.97 months 
Recurrence-free survival  (+): 73 (30.3%); (−): 15 (6.2%); Unknown: 153 (63.5%) 
Follow-up (recurrence-free survival):  47.9± 44.3; 27.9 months 

NOTE: Patient characteristics in the clinical specimen cohort and in the available TCGA dataset for RCC patients. Follow-up information was not available for time to metastasis in the TCGA dataset. Follow-up, mean ± SD and median; survival: (+), survival; (−),death.

miRNA isolation

All specimens were placed in the RNAlater solution (Quiagen) and were stored at −80°C. RNA was extracted from approximately 30 to 50 mg of tissue using the Qiagen RNeasy Kit. miRNA quality was analyzed in Agilent 2100 bioanalyzer.

miRNA profiling

miRNAs were initially profiled from matched normal kidney and tumor specimens from 8 patients with ccRCC. miRNA profiles were compared pair-wise in specimens from individual patients. Four of these patients had low-stage disease: grade 2/stage 1: 3; grade 4/stage 1.5: 1. The remaining 4 patients had high-stage disease: grade 4/stage 3: 2; grade 2/stage 2: 1; grade 3/stage 3.5: 1. In the second round, miRNAs were profiled from 16 additional specimens: 4 normal kidney and 12 tumor specimens (ccRCC: n = 10; papillary: n = 1; chromophobe: n = 1). Among the tumor specimens, seven were low-stage (grade 1, 2/stage 1) and five were high-stage (grade 4/stage 2: 1; grade 3, 4/stage 3.5: 4). Both miRNA profiling studies were performed using a two-color custom-manufactured Agilent 8 × 15K microarray by Dharmacon (Horizon Discovery Group Co). In individual patient comparisons, for each patient sample pair, profiles were filtered, keeping miRNA probes with P value ⇐ 0.05 in at least 1 of the 2 experiments. In the second comparison, data from all specimens (n = 32 total; normal, n = 12; tumor, n = 20) were pooled and analyzed by ANOVA with false detection rate multiple test correction across the tissue type (normal, low stage, or high stage). The processed data were used for agglomerative hierarchical clustering using cosine correlation distance metric. Sample profiles in each comparison (e.g., normal vs. tumor) were subjected to statistical filtering, keeping miRNA probes with P value < 0.05 in at least 8 of the 32 profiled specimens. This resulted in 450 miRNAs for analysis.

miRNA qPCR

Total tissue miRNA preparations were subjected to reverse transcription using RT² miRNA First Strand Kit, and qPCR was performed for each miRNA using the RT² miRNA PCR Array System (SA Biosciences; Qiagen). In addition to the differentially expressed miRNAs, miRNAs that showed no alteration in expression among normal and RCC specimens in the miRNA profiling data were selected for validation as a reference miRNA for normalization. In PCR assays, miR-23a levels consistently did not change among normal and RCC specimens. Therefore, miRNA expression was normalized to miR-23a. Relative expression of individual miRNAs in each specimen was calculated as: (1/2Δcq × 100); ΔCq = Cq(test miR) − Cq(miR-23a) (38, 39). Each specimen was analyzed in duplicate in each miRNA assay. Average normalized value from each specimen was used when computing the data for each miRNA for the specimen cohort.

Statistical analyses

Differences in the levels of the miRNAs that were evaluated by qPCR among kidney tissues (e.g., normal vs. ccRCC, ccRCC vs. oncocytoma) were compared using the Mann–Whitney U test, because the data showed a nonnormal distribution. Similarly, differences in the levels of miRNAs between RCC tissues, with respect to tumor size, grade, or metastasis, were also compared using the Mann–Whitney U test; P values were two-tailed. The Grubbs test (extreme studentized deviate methods) by GraphPad software (https://graphpad.com/quickcalcs/Grubbs1.cfm) was used to identify a significant outlier in a dataset. The levels of the combined miRNA biomarkers (e.g., miR-21+142-5p+194) for each study subject were calculated as follows: [intercept + (α1 × (miR-21)1) + (α2 × (miR-142-5p)1) + (α3 × (miR-194)1)]; α1, α12, and α3: miR-21, miR-142-5p, and miR-194 coefficients, respectively; (miR-21)1, (miR-142-5p)1, and (miR-194)1: the levels of respective miRNAs in subject #1, respectively. The intercept and coefficients for each marker were computed by simultaneously analyzing two or three markers in the logistic regression model (Supplementary Table S1). Logistic regression single-parameter model (i.e., univariate analysis) was used to determine the association of clinical parameters, and the miRNA levels with metastasis, disease-specific mortality, or overall survival. Based on the Youden index (J; ref. 40) from an ROC curve, optimal cut-off values were calculated to compute sensitivity and specificity. Cox-proportional hazards model (i.e., multivariate analysis) was used to determine which of the demographic and pathologic parameters and/or miRNA levels are significant in predicting metastasis (clinical cohort) or the overall survival (TCGA dataset). Stratified Kaplan–Meier plots were prepared for combined markers since these significantly predicted either metastasis or overall survival in both univariate and multivariate analyses.

Differential expression of miRNAs in normal and RCC tissues

We initially performed miRNA profiling on matched normal kidney and tumor specimens from 8 patients with ccRCC. In individual pairwise comparisons, 47 miRNAs were significantly downregulated and 67 were upregulated in normal kidney tissues, when compared with tumor specimens in 7 patients; 46 miRNAs were downregulated and 67 were upregulated in 1 patient. miRNA profiling studies were extended to include sixteen additional specimens. A comparison of the miRNA profiles in a total of 32 specimens showed that although 76 miRNAs were differentially expressed between normal kidney and low-stage tumor tissues, 91 miRNAs were differentially expressed between normal kidney and high-stage tumors; several of these miRNAs were aberrantly expressed in both low- and high-stage tumors. A pairwise comparison of miRNA profiles in the matched specimens and the analysis of miRNA profiles in all 32 specimens showed that 12 miRNAs were consistently and significantly differentially expressed in normal and tumor specimens (Supplementary Table S2). These miRNAs were also among the 20 most differentially expressed miRNAs (cut-off, 2.5-fold difference), based on fold change. qPCR assays were performed on these 12 miRNAs in 5 low-stage, 5 high-stage, and 4 normal specimens that were included in the cohort of 32 specimens. Results showed that seven miRNAs were most significantly differentially expressed in RCC specimens, when compared with normal kidney tissues. These were miR-21, miR-142-3p, miR-142-5p, miR-150, miR-155, miR-192, and miR-194.

Differential expression of miRNAs in RCC subtypes

qPCR assays were performed on 59 normal kidney and 54 tumor specimens from RCC patients. All 7 miRNAs were 2- to 100-fold downregulated in oncocytomas when compared with normal kidney tissues, and these differences were statistically significant (Fig. 1A–G). A comparison between normal kidney tissues and RCC subtypes revealed that although miR-21, miR-155, and/or miR-142-5p were significantly (2–4-fold) upregulated, miR-192 and miR-194 were significantly (3- to >5-fold) downregulated in ccRCC, chromophobe, or papillary tumors, or in tumors with sarcomatoid features (Fig. 1A, E–G). When compared with oncocytoma, miR-155 was 10- to 30-fold upregulated in all RCC subtypes; miR-21, miR-142-3p, and miR-142-5p were significantly upregulated in ccRCC and sarcomatoid RCC; and miR-150 was significantly upregulated in chromophobe tumors. Therefore, not only the seven miRNAs were differentially expressed in normal kidney and tumor specimens, but they were also differentially expressed in various RCC subtypes.

Figure 1.

Expression of the seven most differentially expressed miRNAs in normal kidney and RCC specimens. AG, Levels of each miRNA expressed in normal kidney tissues, oncocytoma, and histologic RCC subtypes are shown. Data, mean ± SEM. Statistical significance (P value) of the differential expression of miRNAs in normal, oncocytoma, and RCC specimens shown in A to G is as follows: only significant differences are listed for each comparison. Normal versus oncocytoma: miR-21: 0.0099; miR-142-3p: P = 0.0122; miR-142-5p: P = 0.0049; miR-150: P = 0.02; miR-155: P = 0.0092; miR-192 and miR-194: P = 0.0004. Normal versus clear cell RCC: miR-21: P = 0.0002; miR-142-5p: P = 0.0338; miR-155: P = 0.0463; miR-192 and miR-194: P < 0.0001. Normal versus papillary: miR-192 and miR-194: P < 0.0001. Normal versus chromophobe: miR-150: P = 0.0317; miR-192: P = 0.0028; miR-194: P = 0.0017. Normal versus sarcomatoid: miR-21: P = 0.002; miR-142-5p: P = 0.0005; miR-192: P = 0.0006; miR-194: P = 0.0004. Oncocytoma versus clear cell RCC: miR-21: P = 0.0013; miR-142-3p: P = 0.0358; miR-142-5p: P = 0.0089; miR-155: P = 0.0107. Oncocytoma versus papillary: miR-155: 0.005. Oncocytoma versus chromophobe: miR-150: P = 0.0317; mir-155: 0.0317. Oncocytoma versus sarcomatoid: miR-21: 0.0043; miR-142-3p: P = 0.0381; miR-142-5p: P = 0.0095.

Figure 1.

Expression of the seven most differentially expressed miRNAs in normal kidney and RCC specimens. AG, Levels of each miRNA expressed in normal kidney tissues, oncocytoma, and histologic RCC subtypes are shown. Data, mean ± SEM. Statistical significance (P value) of the differential expression of miRNAs in normal, oncocytoma, and RCC specimens shown in A to G is as follows: only significant differences are listed for each comparison. Normal versus oncocytoma: miR-21: 0.0099; miR-142-3p: P = 0.0122; miR-142-5p: P = 0.0049; miR-150: P = 0.02; miR-155: P = 0.0092; miR-192 and miR-194: P = 0.0004. Normal versus clear cell RCC: miR-21: P = 0.0002; miR-142-5p: P = 0.0338; miR-155: P = 0.0463; miR-192 and miR-194: P < 0.0001. Normal versus papillary: miR-192 and miR-194: P < 0.0001. Normal versus chromophobe: miR-150: P = 0.0317; miR-192: P = 0.0028; miR-194: P = 0.0017. Normal versus sarcomatoid: miR-21: P = 0.002; miR-142-5p: P = 0.0005; miR-192: P = 0.0006; miR-194: P = 0.0004. Oncocytoma versus clear cell RCC: miR-21: P = 0.0013; miR-142-3p: P = 0.0358; miR-142-5p: P = 0.0089; miR-155: P = 0.0107. Oncocytoma versus papillary: miR-155: 0.005. Oncocytoma versus chromophobe: miR-150: P = 0.0317; mir-155: 0.0317. Oncocytoma versus sarcomatoid: miR-21: 0.0043; miR-142-3p: P = 0.0381; miR-142-5p: P = 0.0095.

Close modal

Differential expression of miRNAs with respect to demographic and pathologic parameters

The aberrant expression of none of the miRNAs significantly correlated with age or gender (P > 0.05). miR-155 levels were significantly elevated in RCC subtypes with tumor size < 4 cm when compared with oncocytoma, and only the levels of miR-194 were significantly different, i.e., downregulated, in high-grade tumors when compared with low-grade tumors (Fig. 2A and B). Similarly, only the levels of miR-142-5p were significantly elevated in high-stage tumors (stage ≥ 2), when compared with low-stage tumors (Supplementary Fig. S1). miR-142-5p levels were also significantly upregulated in RCC specimens from patients with lymph node–positive disease (Fig. 2C).

Figure 2.

Association of miRNA expression with tumor size, grade, and lymph node status. Data on the levels (mean ± SEM) of each miRNA were stratified based on tumor size, tumor type, grade, and lymph node status, and the statistical significance was analyzed by the Mann–Whitney test. In the figure, only those miRNAs that were significantly different between the two parameters are shown. A, Comparison of miRNA levels between oncocytoma (n = 5; size, 4.0 ± 1.0; median, 4.2 cm) and small RCC tumors (<4 cm; n = 12); miR-155 levels were significantly different between these two categories. B, Stratification of miRNA data with respect to tumor grade; miR-194 levels were significantly different between low-grade (<2) and high-grade (≥2) tumors. C, Stratification of miRNA levels in RCC specimens with respect of lymph node invasion; miR-142-5p levels were significantly different among patients with or without lymph node–positive disease at the time of surgery. DF, Correlation of miRNA levels with metastasis. miRNA levels (mean ± SEM) in RCC specimens were compared between patients who developed (+) and those who did not develop (−) metastasis during follow-up. In the figures, only those miRNAs that were significantly different between the two parameters are shown. P values: Manny–Whitney U test.

Figure 2.

Association of miRNA expression with tumor size, grade, and lymph node status. Data on the levels (mean ± SEM) of each miRNA were stratified based on tumor size, tumor type, grade, and lymph node status, and the statistical significance was analyzed by the Mann–Whitney test. In the figure, only those miRNAs that were significantly different between the two parameters are shown. A, Comparison of miRNA levels between oncocytoma (n = 5; size, 4.0 ± 1.0; median, 4.2 cm) and small RCC tumors (<4 cm; n = 12); miR-155 levels were significantly different between these two categories. B, Stratification of miRNA data with respect to tumor grade; miR-194 levels were significantly different between low-grade (<2) and high-grade (≥2) tumors. C, Stratification of miRNA levels in RCC specimens with respect of lymph node invasion; miR-142-5p levels were significantly different among patients with or without lymph node–positive disease at the time of surgery. DF, Correlation of miRNA levels with metastasis. miRNA levels (mean ± SEM) in RCC specimens were compared between patients who developed (+) and those who did not develop (−) metastasis during follow-up. In the figures, only those miRNAs that were significantly different between the two parameters are shown. P values: Manny–Whitney U test.

Close modal

Association of miRNA levels with metastasis

Among the seven most aberrantly expressed miRNAs, miR-21 and miR-142-5p levels were 2- to 3.5-fold upregulated and miR-194 levels were 9.5-fold downregulated in RCC specimens from patients who had metastasis (Fig. 2D–F). Univariate analysis showed that among the demographic and clinical parameters, only higher stage and lymph node positivity significantly correlated with metastasis (Table 2). miR-21 and miR-142-5p positively and miR-194 negatively correlated with metastasis (Table 2). In addition, miRNA combinations, miR-21+194 and miR-21+142-5p+194, significantly but negatively correlated with metastasis, i.e., higher the combined marker levels, the lower is the OR for metastatic disease (Table 2). Although the information on disease-specific mortality was available only on 23 patients/RCC specimens, renal vein invasion, miR-142-5p, and the combined miRNA marker (miR-21+152-5p+194) levels significantly correlated with disease-specific mortality (Table 2).

Table 2.

Determination of the association between metastasis or overall survival and demographic, clinical parameters, or miRNA levels

Clinical specimen cohortTCGA data
MetastasisMetastasis
Markerχ2POR (95% CI)χ2POR (95% CI)
Age 0.15 0.7 0.15 0.5 0.48 NS 
Grade 0.0 0.99 0.0 22.51 <0.0001a 3.46 (2.11–5.93) 
Stage 4.86 0.0274a 4.89 (1.31–23.9) 26.49 <0.0001a 2.97 (2.00–4.63) 
LN (+/-) 10.15 <0.0001a 52 (6.3–1,190) 2.36 0.13 NS 
Gender 2.33 0.127 NS 0.15 0.695 NS 
Tumor size 1.88 0.17 1.88 NA NA NA 
Renal vein 2.4 0.121 NS NA NA NA 
Karnofsky score 0.94 0.33 NS NA NA NA 
miR-21 4.52 0.0334a 1.05 (1.01–1.1) 10.29 0.0013a 2.26 (1.39–3.79) 
miR-142-5p 10.5 0.0012a 1.26 (1.11–1.47) 6.5 0.0108a 1.59 (1.12–2.3) 
miR-194 4.02 0.045a 0.82 (0.95–0.64) 2.19 0.816 NS 
miR-21+194 7.14 0.0075a 0.53 (0.63–0.16) 9.02 0.0027a 0.44 (0.74–0.25) 
miR-21+142-5p+194 9.86 0.0017a 0.53 (0.75–0.33) 9.79 0.0018a 0.43 (0.72–0.25) 
 Disease-specific mortality Overall survival 
Marker χ2 P OR (95% CI) χ2 P OR (95% CI) 
Age 0.0 0.989 NS 8.11 0.0044 1.03 (1.00–1.06) 
Grade 0.0 0.978 NS 39.72 <0.0001a 4.59 (2.92–7.59) 
Stage 1.74 0.187 NS 23.69 <0.0001a 2.05 (1.55–2.76) 
LN (+/−) 0.0 0.997 NS 3.05 0.081 NS 
Gender 1.09 0.297 NS 0.27 0.603 NS 
Tumor size 0.17 0.68 NS NA NA NA 
Renal vein 3.9 0.0483a 12.0 (1.31–277) NA NA NA 
Karnofsky score 0.25 0.614 NS NA NA NA 
miR-21 2.09 0.148 NS 15.77 <0.0001a 2.3 (1.53–3.53) 
miR-142-5p 3.95 0.0468a 1.22 (1.03–1.53) 4.84 0.0278a 1.36 (1.04–1.81) 
miR-194 2.32 0.128 NS 2.19 0.139 NS 
miR-21+194 2.18 0.14 NS 17.46 <0.0001a 0.367 (0.58–0.23) 
miR-21+142-5p+194 5.58 0.0181a 0.37 (0.72–0.13) 17.93 <0.0001a 0.37 (0.58–0.23) 
Clinical specimen cohortTCGA data
MetastasisMetastasis
Markerχ2POR (95% CI)χ2POR (95% CI)
Age 0.15 0.7 0.15 0.5 0.48 NS 
Grade 0.0 0.99 0.0 22.51 <0.0001a 3.46 (2.11–5.93) 
Stage 4.86 0.0274a 4.89 (1.31–23.9) 26.49 <0.0001a 2.97 (2.00–4.63) 
LN (+/-) 10.15 <0.0001a 52 (6.3–1,190) 2.36 0.13 NS 
Gender 2.33 0.127 NS 0.15 0.695 NS 
Tumor size 1.88 0.17 1.88 NA NA NA 
Renal vein 2.4 0.121 NS NA NA NA 
Karnofsky score 0.94 0.33 NS NA NA NA 
miR-21 4.52 0.0334a 1.05 (1.01–1.1) 10.29 0.0013a 2.26 (1.39–3.79) 
miR-142-5p 10.5 0.0012a 1.26 (1.11–1.47) 6.5 0.0108a 1.59 (1.12–2.3) 
miR-194 4.02 0.045a 0.82 (0.95–0.64) 2.19 0.816 NS 
miR-21+194 7.14 0.0075a 0.53 (0.63–0.16) 9.02 0.0027a 0.44 (0.74–0.25) 
miR-21+142-5p+194 9.86 0.0017a 0.53 (0.75–0.33) 9.79 0.0018a 0.43 (0.72–0.25) 
 Disease-specific mortality Overall survival 
Marker χ2 P OR (95% CI) χ2 P OR (95% CI) 
Age 0.0 0.989 NS 8.11 0.0044 1.03 (1.00–1.06) 
Grade 0.0 0.978 NS 39.72 <0.0001a 4.59 (2.92–7.59) 
Stage 1.74 0.187 NS 23.69 <0.0001a 2.05 (1.55–2.76) 
LN (+/−) 0.0 0.997 NS 3.05 0.081 NS 
Gender 1.09 0.297 NS 0.27 0.603 NS 
Tumor size 0.17 0.68 NS NA NA NA 
Renal vein 3.9 0.0483a 12.0 (1.31–277) NA NA NA 
Karnofsky score 0.25 0.614 NS NA NA NA 
miR-21 2.09 0.148 NS 15.77 <0.0001a 2.3 (1.53–3.53) 
miR-142-5p 3.95 0.0468a 1.22 (1.03–1.53) 4.84 0.0278a 1.36 (1.04–1.81) 
miR-194 2.32 0.128 NS 2.19 0.139 NS 
miR-21+194 2.18 0.14 NS 17.46 <0.0001a 0.367 (0.58–0.23) 
miR-21+142-5p+194 5.58 0.0181a 0.37 (0.72–0.13) 17.93 <0.0001a 0.37 (0.58–0.23) 

NOTE: Logistic regression analysis was used to correlate the expression of demographic and clinical parameters as well as individual miRNA levels with metastasis, disease-specific mortality, or overall survival. P vales are two-tailed. For combined markers, intercepts and coefficients are provided in Supplementary Table S1.

aSignificant parameter.

In the multivariate analysis, only the miRNA combinations (i.e., miR-21+194; miR-21+142-5p+194) significantly predicted metastasis (Table 3). Although in this cohort, only 15 patients were positive for metastasis, the two miRNA combination (miR-2+194) had 80% sensitivity and 97.5% specificity, whereas the three-miRNA combination showed 86.7% sensitivity and 82% specificity for predicting metastasis. Kaplan–Meier plots showed that lower levels of miRNA combinations (miR-21+194; miR-21+142-5p+194) significantly stratified the cohort into higher risk for metastasis (Fig. 3A and B).

Table 3.

Multivariate analyses of demographic and clinical parameters and miRNA levels to predict metastasis or overall survival

MetastasisOverall survival
Clinical specimen cohortTCGA dataset
Markerχ2PRR (95% CI)Markerχ2PRR (95% CI)
Age 0.0508 0.822 NS Age 2.42 0.12 NS 
Grade (high/low) 0.549 0.46 NS Grade 2.95 0.0842 NS 
Stage (<T2; ≥T2) 0.4817 0.489 NS Stage 0.574 0.449 NS 
LN 0.191 0.662 NS LN 4.42 0.0355a 3.78 (1.1–11.4) 
Gender 0.072 0.788 NS Gender 0.339 0.56 NS 
Renal vein 0.811 0.368 NS miR-21 4.12 0.042a 1.74 (1.01–3.08) 
Tumor size (<4 cm; ≥4 cm) 0.31 0.578 NS miR-194 6.37 0.0116a 0.735 (0.93–0.6) 
miR-21+194 5.73 0.0166a 0.5 (0.97–0.2) miR-21+194 7.29 0.0069a 0.43 (0.79–0.22) 
miR-21+142-5p+ 194 5.48 0.0193a 0.54 (0.93–0.24) miR-21+142-5p+194 5.47 0.0193a 0.49 (0.89–0.26) 
MetastasisOverall survival
Clinical specimen cohortTCGA dataset
Markerχ2PRR (95% CI)Markerχ2PRR (95% CI)
Age 0.0508 0.822 NS Age 2.42 0.12 NS 
Grade (high/low) 0.549 0.46 NS Grade 2.95 0.0842 NS 
Stage (<T2; ≥T2) 0.4817 0.489 NS Stage 0.574 0.449 NS 
LN 0.191 0.662 NS LN 4.42 0.0355a 3.78 (1.1–11.4) 
Gender 0.072 0.788 NS Gender 0.339 0.56 NS 
Renal vein 0.811 0.368 NS miR-21 4.12 0.042a 1.74 (1.01–3.08) 
Tumor size (<4 cm; ≥4 cm) 0.31 0.578 NS miR-194 6.37 0.0116a 0.735 (0.93–0.6) 
miR-21+194 5.73 0.0166a 0.5 (0.97–0.2) miR-21+194 7.29 0.0069a 0.43 (0.79–0.22) 
miR-21+142-5p+ 194 5.48 0.0193a 0.54 (0.93–0.24) miR-21+142-5p+194 5.47 0.0193a 0.49 (0.89–0.26) 

NOTE: Cox proportional hazards analysis was performed by including demographic and clinical parameters and the levels of each miRNA. Inclusion of all miRNAs in the model simultaneously, along with clinical parameters, resulted in an unstable model. Multivariate analysis using the Cox proportional hazards model could not be performed for the TCGA dataset since the follow-up on time to metastasis was not available. miRNAs that reached significance are shown. χ2 and P shown are for the effect of Wald test.

aSignificant parameter.

Figure 3.

Kaplan–Meier plots for metastasis and overall survival based on miRNA levels. A and B, Data on miRNA levels in RCC specimens (clinical specimen cohort) were stratified as high and low expression based on the Youden index from an ROC curve that was generated from the logistic regression (univariate) analysis. Stratified data were then used to generate Kaplan–Meier plots with respect to metastasis (A, B). C and D, Data on miRNA levels in the TCGA dataset were stratified as high and low expression based on the Youden index obtained from an ROC curve that was generated from the logistic regression (univariate) analysis. Stratified data in were then used for generating Kaplan–Meier plots with respect to overall survival.

Figure 3.

Kaplan–Meier plots for metastasis and overall survival based on miRNA levels. A and B, Data on miRNA levels in RCC specimens (clinical specimen cohort) were stratified as high and low expression based on the Youden index from an ROC curve that was generated from the logistic regression (univariate) analysis. Stratified data were then used to generate Kaplan–Meier plots with respect to metastasis (A, B). C and D, Data on miRNA levels in the TCGA dataset were stratified as high and low expression based on the Youden index obtained from an ROC curve that was generated from the logistic regression (univariate) analysis. Stratified data in were then used for generating Kaplan–Meier plots with respect to overall survival.

Close modal

Association of miRNA levels with metastasis and overall survival in a TCGA dataset

We next analyzed whether the seven miRNAs identified as significantly differentially expressed in RCC can be validated in a TCGA RCC dataset. This dataset contains transcript expression data on tumor specimens from 241 RCC patients (Table 1). In univariate analysis, miR-21, miR-142-5p, and the miRNA combinations (miR-2+194; miR-21+142-5p+194) significantly associated with metastasis and overall survival (Table 2). In multivariate analysis, lymph node metastasis and the miRNA combinations (miR-21+194; miR-21+142-5p+194) were independent predictors of overall survival (Table 3). Kaplan–Meier plots showed that lower levels of miRNA combinations (miR-21+194; miR-21+142-5p+194) significantly stratified the cohort into higher risk for death (Fig. 3C and D). The combination of miR-142-5p either with miR-21 or miR-194 did not consistently significantly correlate with metastasis, disease-specific mortality, and overall survival in the clinical and TCGA cohorts.

Molecular subtyping of kidney tumors is clinically significant from the standpoint of differentiating benign oncocytomas from other malignant histologic subtypes and identifying metastasis or predicting it at the time of diagnosis. Although the aberrant expression of miRNAs, in general, and in RCC, in particular, has been useful for understanding the oncogenic networks that promote disease progression, an miRNA signature(s) has not been reduced to clinical practice as a diagnostic or prognostic marker. A likely reason for this is the lack of uniformity among different studies regarding the identification of clinically significant miRNAs; most miRNAs identified as prognostic indicators have been reported in a single or at the most two studies (19, 20, 26).

The discrepancy in various studies may be due to the use of different specimen cohorts in different studies; more importantly however, if the discovery tools such as miRNA profiling are not applied to normal and tumor tissues from the same patients, comorbid conditions and genetic differences among patient cohorts, which are unrelated to RCC, may substantially influence the results. In our study, miRNA profiling was initially performed on matched normal kidney and RCC tissues from the same patients, and the profiling was then extended to unmatched normal kidney and RCC specimens, the latter adding rigor to the discovery phase. It is noteworthy that although previous studies have identified miRNAs such as miR-126b, miR-200c, miR-129-3p, miR-23b, miR-27b, miR-99a and miR-1826 as significantly aberrantly expressed miRNAs in RCC, in our study, these miRNAs were found to be either not differentially expressed or were not in the top 20 list (19, 20, 26).

In our study, PCR assays for all uniquely differentially expressed miRNAs defined an aberrantly expressed miRNA signature of seven miRNAs—miR-21, miR-142-3p, miR-142-5p, miR-150, miR-155, miR-192, and miR-194. These seven miRNAs were confirmed by PCR assays conducted in the entire clinical specimen cohort of 113 specimens and then validated in a TCGA dataset. This interconnected four-pronged approach of miRNA profiling, matched and unmatched specimens, PCR assays, and the analysis of TCGA dataset further narrowed the signature to 3 miRNAs as a possible prognosticator for disease outcome.

Among the seven miRNAs identified in this study as most aberrantly expressed in RCC specimens, miR-21, miR-142-3p, miR-142-5p, and miR-155 have been shown to promote oncogenic functions and miR-192 and miR-194 as having tumor-suppressive functions in RCC (25, 31, 34, 35, 41–45). miR-21 has been previously identified as an oncogenic miRNA and has been shown to promote tumor growth and epithelial–mesenchymal transition in RCC cells. Furthermore, miR-21 upregulates AKT-mTORC1 and JAK2/Stat3 axis through the downregulation of tumor suppressor PDCD4 (35, 43, 46–48). miR-142-5p has been shown to promote proliferation and migration of RCC cells by targeting BTG3, but it has not been correlated with clinical outcome (45). miR-194 has been shown to decrease cell migration and invasion, and in one study, higher levels of miR-194 were shown to correlate with longer cancer-specific and overall survival in RCC patients (30, 42). However, in none of the studies, the three miRNAs (i.e., miR-21, miR-142-5p, and miR-194) have been identified as being differentially expressed, as a group, or their combination as being potentially significant in predicting disease outcome.

Identification of small renal mass from oncocytoma is clinically important. Among the seven miRNAs, only the expression of miR-155 was significantly different between oncocytoma and small RCC tumors. The main finding of this study, however, is the miRNA combination signatures, miR-21+194 and miR-21+142-5p+194, that may be potentially accurate in predicting metastasis and disease-specific mortality. A limitation of our study is that in the clinical cohort, data on disease-specific mortality were available only in a few patients. Another limitation of the study is that in the TCGA dataset, no follow-up data were available on the time for metastasis or disease-specific mortality, and insufficient data are available on recurrence-free survival. Therefore, evaluation of the independent prognostic potential of any parameter or biomarker to predict disease outcome in the TCGA dataset could not be performed.

In summary, our study has used a discovery and multiple validation approaches to identify aberrantly expressed miRNAs in RCC. Based on miRNA profiling, PCR confirmation, and TCGA dataset validation, our study identified a three-miRNA signature that shows consistent aberrant expression in RCC and is potentially clinically significant in predicting disease outcome.

No potential conflicts of interest were disclosed.

Conception and design: S.D. Lokeshwar, T.J Yates, V.B. Lokeshwar

Development of methodology: S.D. Lokeshwar, T.J Yates, Z. Klaassen

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S.D. Lokeshwar, A. Talukder, T.J Yates, M. Garcia-Roig, S.S. Lahorewala, B.R. Kava, M. Manoharan, M.S. Soloway

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S.D. Lokeshwar, A. Talukder, T.J Yates, M.J.P. Hennig, M. Garcia-Roig

Writing, review, and/or revision of the manuscript: S.D. Lokeshwar, T.J Yates, M.J.P. Hennig, M. Garcia-Roig, N.N. Mullani, Z. Klaassen, B.R. Kava, M. Manoharan, V.B. Lokeshwar

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S.D. Lokeshwar, M.J.P. Hennig, M. Manoharan, V.B. Lokeshwar

Study supervision: V.B. Lokeshwar

This work was supported by the NIH (grant #: 5R01CA176691-03, to V.B. Lokeshwar) and the Medical College of Georgia, Augusta University funds. M.J.P. Hennig was a fellow of the Biomedical Exchange Program, International Academy of Life Sciences. A major part of this work was conducted in the corresponding author's prior institution, Department of Urology, University of Miami Miller School of Medicine, Miami, Florida.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Gomez-Dantes
H
,
Fullman
N
,
Lamadrid-Figueroa
H
,
Cahuana-Hurtado
L
,
Darney
B
,
Avila-Burgos
L
, et al
Dissonant health transition in the states of Mexico, 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013
.
Lancet
2016
;
388
:
2386
402
.
2.
Torre
LA
,
Siegel
RL
,
Ward
EM
,
Jemal
A
. 
Global cancer incidence and mortality rates and trends–an update
.
Cancer Epidemiol Biomarkers Prev
2016
;
25
:
16
27
.
3.
Hsieh
JJ
,
Purdue
MP
,
Signoretti
S
,
Swanton
C
,
Albiges
L
,
Schmidinger
M
, et al
Renal cell carcinoma
.
Nat Rev Dis Primers
2017
;
3
:
17009
.
4.
Wu
J
,
Zhang
P
,
Zhang
G
,
Wang
H
,
Gu
W
,
Dai
B
, et al
Renal cell carcinoma histological subtype distribution differs by age, gender, and tumor size in coastal Chinese patients
.
Oncotarget
2017
;
8
:
71797
804
.
5.
Carlo
MI
,
Chaim
J
,
Patil
S
,
Kemel
Y
,
Schram
AM
,
Woo
K
, et al
Genomic characterization of renal medullary carcinoma and treatment outcomes
.
Clin Genitourin Cancer
2017
;
15
:
e987
94
.
6.
Casuscelli
J
,
Weinhold
N
,
Gundem
G
,
Wang
L
,
Zabor
EC
,
Drill
E
, et al
Genomic landscape and evolution of metastatic chromophobe renal cell carcinoma
.
JCI Insight
2017
;
2
.
7.
Hakimi
AA
,
Reznik
E
,
Lee
CH
,
Creighton
CJ
,
Brannon
AR
,
Luna
A
, et al
An integrated metabolic atlas of clear cell renal cell carcinoma
.
Cancer Cell
2016
;
29
:
104
16
.
8.
Li
Z
,
Chen
Y
,
Hu
S
,
Zhang
J
,
Wu
J
,
Ren
W
, et al
Integrative analysis of protein-coding and non-coding RNAs identifies clinically relevant subtypes of clear cell renal cell carcinoma
.
Oncotarget
2016
;
7
:
82671
85
.
9.
Eckel-Passow
JE
,
Igel
DA
,
Serie
DJ
,
Joseph
RW
,
Ho
TH
,
Cheville
JC
, et al
Assessing the clinical use of clear cell renal cell carcinoma molecular subtypes identified by RNA expression analysis
.
Urol Oncol
2015
;
33
:
68
.
e17–23
.
10.
Wang
L
,
Williamson
SR
,
Wang
M
,
Davidson
DD
,
Zhang
S
,
Baldridge
LA
, et al
Molecular subtyping of metastatic renal cell carcinoma: implications for targeted therapy
.
Mol Cancer
2014
;
13
:
39
.
11.
Kutikov
A
,
Smaldone
MC
. 
The small renal mass and its management in urologic practice
.
Urol Clin North Am
2017
;
44
:
xvii
.
12.
Tan
PH
,
Cheng
L
,
Rioux-Leclercq
N
,
Merino
MJ
,
Netto
G
,
Reuter
VE
, et al
Renal tumors: diagnostic and prognostic biomarkers
.
Am J Surg Pathol
2013
;
37
:
1518
31
.
13.
Taneja
SS
. 
Small renal mass
.
Urol Clin North Am
2017
;
44
:
xv
.
14.
Tang
DH
,
Nawlo
J
,
Chipollini
J
,
Gilbert
SM
,
Poch
M
,
Pow-Sang
JM
, et al
Management of renal masses in an octogenarian cohort: Is there a right approach?
Clin Genitourin Cancer
2017
;
15
:
696
703
.
15.
Choueiri
TK
,
Motzer
RJ
. 
Systemic therapy for metastatic renal-cell carcinoma
.
N Engl J Med
2017
;
376
:
354
66
.
16.
Ho
TH
,
Jonasch
E
. 
Genetic kidney cancer syndromes
.
J Natl Compr Canc Netw
2014
;
12
:
1347
55
.
17.
Randall
JM
,
Millard
F
,
Kurzrock
R
. 
Molecular aberrations, targeted therapy, and renal cell carcinoma: current state-of-the-art
.
Cancer Metastasis Rev
2014
;
33
:
1109
24
.
18.
Tennenbaum
DM
,
Manley
BJ
,
Zabor
E
,
Becerra
MF
,
Carlo
MI
,
Casuscelli
J
, et al
Genomic alterations as predictors of survival among patients within a combined cohort with clear cell renal cell carcinoma undergoing cytoreductive nephrectomy
.
Urol Oncol
2017
;
35
:
532.e7
532.e13
.
19.
Gu
L
,
Li
H
,
Chen
L
,
Ma
X
,
Gao
Y
,
Li
X
, et al
MicroRNAs as prognostic molecular signatures in renal cell carcinoma: a systematic review and meta-analysis
.
Oncotarget
2015
;
6
:
32545
60
.
20.
Kurozumi
A
,
Goto
Y
,
Okato
A
,
Ichikawa
T
,
Seki
N
. 
Aberrantly expressed microRNAs in bladder cancer and renal cell carcinoma
.
J Hum Genet
2017
;
62
:
49
56
.
21.
Ling
H
,
Girnita
L
,
Buda
O
,
Calin
GA
. 
Non-coding RNAs: the cancer genome dark matter that matters!
Clin Chem Lab Med
2017
;
55
:
705
14
.
22.
Morais
M
,
Dias
F
,
Teixeira
AL
,
Medeiros
R
. 
MicroRNAs and altered metabolism of clear cell renal cell carcinoma: potential role as aerobic glycolysis biomarkers
.
Biochim Biophys Acta
2017
;
1861
:
2175
85
.
23.
Sellitti
DF
,
Doi
SQ
. 
MicroRNAs in renal cell carcinoma
.
Microrna
2015
;
4
:
26
35
.
24.
Kowalik
CG
,
Palmer
DA
,
Sullivan
TB
,
Teebagy
PA
,
Dugan
JM
,
Libertino
JA
, et al
Profiling microRNA from nephrectomy and biopsy specimens: predictors of progression and survival in clear cell renal cell carcinoma
.
BJU Int
2017
;
120
:
428
40
.
25.
Li
G
,
Zhao
A
,
Peoch
M
,
Cottier
M
,
Mottet
N
. 
Detection of urinary cell-free miR-210 as a potential tool of liquid biopsy for clear cell renal cell carcinoma
.
Urol Oncol
2017
;
35
:
294
9
.
26.
Zhu
J
,
Ma
X
,
Zhang
Y
,
Ni
D
,
Ai
Q
,
Li
H
, et al
Establishment of a miRNA-mRNA regulatory network in metastatic renal cell carcinoma and screening of potential therapeutic targets
.
Tumour Biol
2016 Nov 2
. [Epub ahead of print].
27.
Garcia-Donas
J
,
Beuselinck
B
,
Inglada-Perez
L
,
Grana
O
,
Schoffski
P
,
Wozniak
A
, et al
Deep sequencing reveals microRNAs predictive of antiangiogenic drug response
.
JCI Insight
2016
;
1
:
e86051
.
28.
Machackova
T
,
Mlcochova
H
,
Stanik
M
,
Dolezel
J
,
Fedorko
M
,
Pacik
D
, et al
MiR-429 is linked to metastasis and poor prognosis in renal cell carcinoma by affecting epithelial-mesenchymal transition
.
Tumour Biol
2016
;
37
:
14653
8
.
29.
Sun
X
,
Zhang
J
. 
Dysfunctional miRNA-mediated regulation in chromophobe renal cell carcinoma
.
PLoS One
2016
;
11
:
e0156324
.
30.
Nofech-Mozes
R
,
Khella
HW
,
Scorilas
A
,
Youssef
L
,
Krylov
SN
,
Lianidou
E
, et al
MicroRNA-194 is a marker for good prognosis in clear cell renal cell carcinoma
.
Cancer Med
2016
;
5
:
656
64
.
31.
Rasti
A
,
Mehrazma
M
,
Madjd
Z
,
Keshtkar
AA
,
Roudi
R
,
Babashah
S
. 
Diagnostic and prognostic accuracy of miR-21 in renal cell carcinoma: a systematic review protocol
.
BMJ Open
2016
;
6
:
e009667
.
32.
Petrozza
V
,
Carbone
A
,
Bellissimo
T
,
Porta
N
,
Palleschi
G
,
Pastore
AL
, et al
Oncogenic microRNAs characterization in clear cell renal cell carcinoma
.
Int J Mol Sci
2015
;
16
:
29219
25
.
33.
Fedorko
M
,
Pacik
D
,
Wasserbauer
R
,
Juracek
J
,
Varga
G
,
Ghazal
M
, et al
MicroRNAs in the pathogenesis of renal cell carcinoma and their diagnostic and prognostic utility as cancer biomarkers
.
Int J Biol Markers
2016
;
31
:
e26
37
.
34.
Liu
Z
,
Lu
Y
,
Xiao
Y
,
Lu
Y
. 
Upregulation of miR-21 expression is a valuable predicator of advanced clinicopathological features and poor prognosis in patients with renal cell carcinoma through the p53/p21cyclin E2Bax/caspase-3 signaling pathway
.
Oncol Rep
2017
;
37
:
1437
44
.
35.
Szabo
Z
,
Szegedi
K
,
Gombos
K
,
Mahua
C
,
Flasko
T
,
Harda
K
, et al
Expression of miRNA-21 and miRNA-221 in clear cell renal cell carcinoma (ccRCC) and their possible role in the development of ccRCC
.
Urol Oncol
2016
;
34
:
533
e21–e27
.
36.
Li
Y
,
Chen
D
,
Li
Y
,
Jin
L
,
Liu
J
,
Su
Z
, et al
Identification of miR130b as an oncogene in renal cell carcinoma
.
Mol Med Rep
2016
;
13
:
1902
8
.
37.
Fedorko
M
,
Stanik
M
,
Iliev
R
,
Redova-Lojova
M
,
Machackova
T
,
Svoboda
M
, et al
Combination of MiR-378 and MiR-210 serum levels enables sensitive detection of renal cell carcinoma
.
Int J Mol Sci
2015
;
16
:
23382
9
.
38.
Chi
A
,
Shirodkar
SP
,
Escudero
DO
,
Ekwenna
OO
,
Yates
TJ
,
Ayyathurai
R
, et al
Molecular characterization of kidney cancer: association of hyaluronic acid family with histological subtypes and metastasis
.
Cancer
2012
;
118
:
2394
402
.
39.
Gahan
JC
,
Gosalbez
M
,
Yates
T
,
Young
EE
,
Escudero
DO
,
Chi
A
, et al
Chemokine and chemokine receptor expression in kidney tumors: molecular profiling of histological subtypes and association with metastasis
.
J Urol
2012
;
187
:
827
33
.
40.
Youden
WJ
. 
Index for rating diagnostic tests
.
Cancer
1950
;
3
:
32
5
.
41.
Ji
H
,
Tian
D
,
Zhang
B
,
Zhang
Y
,
Yan
D
,
Wu
S
. 
Overexpression of miR-155 in clear-cell renal cell carcinoma and its oncogenic effect through targeting FOXO3a
.
Exp Ther Med
2017
;
13
:
2286
92
.
42.
Khella
HW
,
Bakhet
M
,
Allo
G
,
Jewett
MA
,
Girgis
AH
,
Latif
A
, et al
miR-192, miR-194 and miR-215: a convergent microRNA network suppressing tumor progression in renal cell carcinoma
.
Carcinogenesis
2013
;
34
:
2231
9
.
43.
Li
X
,
Xin
S
,
He
Z
,
Che
X
,
Wang
J
,
Xiao
X
, et al
MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor PDCD4 and promotes cell transformation, proliferation, and metastasis in renal cell carcinoma
.
Cell Physiol Biochem
2014
;
33
:
1631
42
.
44.
Li
Y
,
Chen
D
,
Jin
LU
,
Liu
J
,
Li
Y
,
Su
Z
, et al
Oncogenic microRNA-142-3p is associated with cellular migration, proliferation and apoptosis in renal cell carcinoma
.
Oncol Lett
2016
;
11
:
1235
41
.
45.
Liu
L
,
Liu
S
,
Duan
Q
,
Chen
L
,
Wu
T
,
Qian
H
, et al
MicroRNA-142-5p promotes cell growth and migration in renal cell carcinoma by targeting BTG3
.
Am J Transl Res
2017
;
9
:
2394
402
.
46.
Bera
A
,
Das
F
,
Ghosh-Choudhury
N
,
Kasinath
BS
,
Abboud
HE
,
Choudhury
GG
. 
microRNA-21-induced dissociation of PDCD4 from rictor contributes to Akt-IKKbeta-mTORC1 axis to regulate renal cancer cell invasion
.
Exp Cell Res
2014
;
328
:
99
117
.
47.
Fritz
HK
,
Lindgren
D
,
Ljungberg
B
,
Axelson
H
,
Dahlback
B
. 
The miR(21/10b) ratio as a prognostic marker in clear cell renal cell carcinoma
.
Eur J Cancer
2014
;
50
:
1758
65
.
48.
Su
Y
,
Zhao
A
,
Cheng
G
,
Xu
J
,
Ji
E
,
Sun
W
. 
The IGF-I/JAK2-STAT3/miR-21 signaling pathway may be associated with human renal cell carcinoma cell growth
.
Cancer Biomark
2017
;
19
:
289
96
.