Skip to main content
Erschienen in: Cancer Cell International 1/2022

Open Access 01.12.2022 | Review

A review on the role of mir-16-5p in the carcinogenesis

verfasst von: Soudeh Ghafouri-Fard, Tayyebeh Khoshbakht, Bashdar Mahmud Hussen, Sara Tharwat Abdullah, Mohammad Taheri, Mohammad Samadian

Erschienen in: Cancer Cell International | Ausgabe 1/2022

Abstract

miR-16-5p is microRNA with important roles in the development of diverse malignancies including neuroblastoma, osteosarcoma, hepatocellular carcinoma, cervical cancer, breast cancer, brain tumors, gastrointestinal cancers, lung cancer and bladder cancer. This miRNA has 22 nucleotides. hsa-miR-16-5p is produced by MIR16-1 gene. First evidence for its participation in the carcinogenesis has been obtained by studies reporting deletion and/or down-regulation of these miRNAs in chronic lymphocytic leukemia. Subsequent studies have shown down-regulation of miR-16-5p in a variety of cancer cell lines and clinical samples. Besides, tumor suppressor role of miR-16-5p has been verified in animal models of different types of cancers. Studies in these models have shown that over-expression of this miRNA or modulation of expression of lncRNAs that sponge this miRNA can block carcinogenic processes. In the current review, we summarize function of miR-16-5p in the development and progression of different cancers.
Hinweise

Publisher’s note

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

Introduction

MicroRNAs (miRNAs) are small-sized transcripts that regulate expression of genes at post-transcriptional level through specific targeting of mRNAs. With sizes about 21–25 nucleotides, miRNAs are originated from coding and non-coding transcription units in introns, exons or intergenic areas [1]. They are produced in a multi-step process involving both nuclear and cytoplasmic proteins. They are involved in the carcinogenic process, since they can regulate expression of several oncogenes and tumor suppressor genes as well as activities of cancer-associated pathways [2]. Expression pattern and function of several miRNAs have been assessed in different cancer types. Since these small-sized transcripts are stable in the circulation or other biofluids, they represent potential biomarkers for diagnostic and follow-up purposes [3]. Dysregulation of miRNAs has been correlated with evolution of cancers, hence they are regarded as molecular tools for non-invasive assessment of cancer occurrence and its prognosis [4].
miR-16-5p is an example of this class of transcripts with important roles in the development of diverse malignancies including neuroblastoma, osteosarcoma, hepatocellular carcinoma, cervical cancer, breast cancer, brain tumors, gastrointestinal cancers, lung cancer and bladder cancer. This miRNA has 22 nucleotides and is present in Homo sapiens. Homo sapiens hsa-miR-16-5p is produced by MIR16-1 gene.
miR-16-1 is allocated at 13q14.3 along with miR-15a. This miRNA cluster is the target of 13q deletions in chronic lymphocytic leukemia (CLL). miRNAs encoded by this locus have tumor suppressor functions. First evidence for its participation in the carcinogenesis has been obtained by studies reporting deletion and/or down-regulation of these miRNAs in (CLL) [5]. The tumor suppressor functions of miR-15a/16 − 1 are exerted through targeting the BCL2 oncogene. Through a high-throughput study in a leukemic cell line model, Colin et al. have found enrichment in AU-rich elements in the elements of the miR-15a/16 − 1 signature [6].
Subsequently, different studies have assessed role of miR-16-5p in the carcinogenesis using in vitro and in vivo techniques. Moreover, expression pattern of miR-16-5p has been evaluated in clinical samples gathered from patients with diverse malignancies. In the current review, we summarize function of miR-16-5p in the development and progression of different cancers using the above-mentioned lines of evidence. The reason for selection of this miRNA in this review article is the important role of this miRNA in the suppression of carcinogenesis, its down-regulation in a variety of solid and hematological malignancies and its potential as an anti-cancer target. The following strategy was used for selection of papers: publication in full-text English language in a peer-reviewed journal and detailed description of conducted methods. In addition, papers should include in vitro functional studies or expression assays in clinical samples.

Cell line studies

Cell line studies have indicated important roles of miR-16-5p in the carcinogenesis. Moreover, these studies have shown the inhibitory effects of this miRNA on transcription of several genes, particularly a number of known oncogenes. An in vitro study in neuroblastoma has shown interaction between miR-15a, miR‐15b and miR‐16 and MYCN transcript. Based on the results of luciferase reporter assay these miRNAs bind with 3’UTR of MYCN transcript leading to suppression of its expression. Forced up-regulation of these miRNAs has decreased proliferative potential, migratory ability, and invasion of neuroblastoma cells [7]. Another study in neuroblastoma has shown that the oncogenic circular RNA circ-CUX1 enhances tumorigenesis of neuroblastoma and their glycolysis through targeting miR-16-5p. Moreover, miR-16-5p tumor suppressor impact has been partially decreased by transfection of circ-CUX1 overexpressing vectors. DMRT2 has been found to be targeted by miR-16-5p in neuroblastoma cells [8].
miR-16-5p has been to be down-regulated in osteosarcoma cell lines compared with control cells, parallel with up-regulation of Smad3. Up-regulation of miR-16-5p has suppressed proliferation, migratory potential and invasive features of osteosarcoma cells and increased the cytotoxic effects of cisplatin on these cells. Moreover, miR-16-5p over-expression has led to reduction of Smad3 expression. Notably, cells harboring Smad3 mutation have not responded to miR-16-5p over-expression, indicating that miR-16-5p suppresses invasive properties of osteosarcoma cells through suppressing expression of Smad3 [9]. miR-16-5p effect in suppression of tetraspanin 15 gene has also been involved in the inhibition of osteosarcoma cells proliferation, migration and invasion [10]. Figure 1 shows tumor suppressor role of miR-16-5p in different types of cancer.
The long non-coding RNA (lncRNA) AGAP2-AS1 which targets miR-16-5p has been shown to be up-regulated in hepatocellular carcinoma cell lines. This lncRNA could promote proliferation, migratory aptitude, invasiveness and epithelial-mesenchymal transition (EMT) of these cells through acting as a sponge for miR-16-5p. ANXA11 has been found as a target of miR-16-5p in hepatocellular carcinoma cells, mediating the impacts of miR-16-5p and AGAP2-AS1 in these cells and enhancing activity of AKT signaling. Notably, hypoxia has been shown to increase levels of AGAP2-AS1 in these cells [11]. Another study has confirmed down-regulation of miR-16-5p in hepatocellular cancer cells. Dual-Luciferase reporter gene assay has validated the regulatory role of miR-16-5p on expression of Insulin like growth factor1 receptor (IGF1R). IGF1R down-regulation has decreased the suppressive role of miR-16- 5p on proliferation ability and metastatic potential of hepatocellular cancer cells [12]. Moreover, down-regulation of miR-16-5p by lncRNA TTN-AS1 has been shown to promote resistance to sorafenib through enhancement of expression of cyclin E1 [13]. Finally, another study in hepatocellular carcinoma has shown that SNHG22 increases tumorigenic ability of cancer cells and their angiogenesis though induction of DNA methylation in miR-16-5p [14].
In cervical cancer cells, miR-16‐5p affects radiosensitivity through regulation of expression of coactivator‐associated arginine methyltransferase 1 [15]. Moreover, it can influence metabolic reprogramming and chemoresistance through regulation of Pyruvate Dehydrogenase Kinase 4 (PDK4) expression [16].
In breast cancer cell, down-regulation of miR-16-5p has been associated with high migratory and proliferative potential of cells, induction of cell cycle progression and reduction of cell apoptosis. miR-16-5p could restrain activity of the Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway and reduce expression of AKT3 gene, thus inhibiting development of breast cancer [17]. miR-16-5p could also suppress proliferation of breast cancer cells through down-regulating expression of ANLN [18]. The inhibitory effect of miR-16 -5p in breast cancer cells proliferation and invasiveness can be mediated through regulation of Vascular Endothelial Growth Factor A (VEGFA) expression [19]. Finally, ATXN8OS has been shown to enhance tamoxifen resistance through sponging miR-16-5p [20].
Moreover, miR-16‐5p has been shown to be commonly down‐regulated in astrocytic gliomas. This miRNA could regulate proliferation and apoptosis of these cells as well as effect of cytotoxic agents on these cells [21]. Another study in glioma cells has shown that TIIA could inhibit viability of cells, their migratory potential and invasiveness, and decrease levels of Cyclin D1, Matrix metallopeptidase 9 (MMP-9) and Vimentin via regulation of miR-16-5p/Talin-1 axis [22].
Summary of studies that evaluated expression of miR-16-5p or its partners in cell lines is presented in Table 1.
Table 1
Expression of miR-16-5p or its partners in cell lines (∆: knock-down or deletion, TIIA: Tanshinone IIA, CuET: diethyldithiocarbamate-copper complex, EPCs: endothelial progenitor cells, RANKL: receptor activator of nuclear factor-κB ligand, IDH: Isocitrate dehydrogenase)
Tumor type
Targets/ Regulators and Signaling Pathways
Cell line
Function
Reference
Neuroblastoma
MYCN
SK-N‐BE(2), NB‐19, and SH‐EP Tet21N
↑↑ miR-16-5p: ↓ proliferation, migration, and invasion
[7]
Circ-CUX1, DMRT2
HUVEC, GI-LI-N, SK-N-SH and IMR-32
∆ Circ-CUX1 (which suppresses miR-16-5p): ↓ proliferation, migration, invasion , and glycolysis
[8]
Osteosarcoma
Smad3
hFOB1.19, MG63, SaOS2, HOS, and U2OS
↑↑ miR-16-5p: ↓ proliferation, migration, invasion, and ↑ therapeutic effect of cisplatin
[9]
TSPAN15, PI3K/AKT signaling pathway
hFOB 1.19, MG63, Saos2 and HOS
↑↑ miR-16-5p: ↓ viability, migration, invasion
[10]
LINC00662, ITPR1
U2OS, SAOS-2, 143B, and MG63, HFOB 1.19
∆ LINC00662 (which sponges miR-16-5p): ↓ proliferation, migration, invasion , and stemness property maintenance
[23]
hsa_circ_0005721, TEP1
hFOB, 143B, U-2OS, HOS and Saos-2
∆ hsa_circ_0005721 (which sponges miR-16-5p): ↓ viability, migration, invasion
[24]
Hepatocellular carcinoma
AGAP2-AS1, ANXA11, AKT signaling
LO2, Hep3B, HCCLM3, Huh7, MHCC-97 H and SMMC-7721
↑↑ AGAP2-AS1: ↑ proliferation, migration, invasion , and ↓ apoptosis
[11]
IGF1R
SMMC-7721, HL-7702
↑↑ miR-16-5p: ↓ proliferation, migration, invasion, and EMT process
[12]
TTN-AS1, cyclin E1, PTEN/Akt signaling pathway
Bel7404 and HepG2
∆ TTN-AS1 (which sponges miR-16-5p): ↓ sorafenib resistance, ↑ apoptosis
∆ miR-16-5p: ↑ sorafenib resistance, ↓ apoptosis
[13]
SNHG22, EZH2, DNMT1
HLE-3, Huh7, HCCLM6, MHCC97H and SNU-398
∆ SNHG22
(which suppresses the transcription of miR-16-5p): ↓ proliferation, invasion, and angiogenesis
∆ miR-16-5p: ↑ proliferation, migration, invasion, and angiogenesis
[14]
Cervical cancer
CARM1
HeLa, C-33 A, CaSki, HeLa229, SiHa, END1/
E6E7
↑↑ miR-16-5p: ↓ colony formation, and radioresistance, ↑ apoptosis
[15]
PDK4
HeLa, SiHa, HeLa/Dox, and SiHa/Dox
↑↑ miR-16-5p: ↓ proliferation, glucose consumption, lactate production, and ATP levels, and resistance to Dox treatment
[16]
DLX6-AS1, ARPP19
End1/E6E7, SiHa, HeLa, C-33 A, and CaSki
∆ DLX6-AS1 (which sponges miR-16-5p): ↓ proliferation, migration, and EMT process, ↑ apoptosis
[25]
Breast cancer
AKT3, NF-κB pathway
BT-549 and MCF-7
↑↑ miR-16-5p: ↓ proliferation, migration, ↑ apoptosis, cell cycle arrest
[17]
ANLN
MCF-7, T47D, MDA-MB-231, EMF-192 A, SKBR-3 and MCF-10 A, HEK293T
↑↑ miR-16-5p: ↓ proliferation, migration, invasion, and ↑ apoptosis, G2/M phase arrest
[18]
VEGFA , Hypoxia-inducible factor-α (HIF-α)
MCF-7 and MDA-MB-231, MDA-MB-435, MDA-MB-468 and T47D, MCF10A
↑↑ miR-16-5p: ↓ proliferation, invasion, colony formation, ↑ apoptosis
[19]
ATXN8OS, VASP
MCF-10 A MCF-7, and BT-549
∆ ATXN8OS (which sponges miR-16-5p): ↑ tamoxifen sensitivity
[20]
Gliomas
WEE1,
CHEK1 and MCL1
A172, T98G, U251MG, U138MG and U87MG, TP365MG
↑↑ miR-16-5p: ↓ proliferation, viability, ↑ apoptosis, cell cycle arrest, response to irradiation and
chemotherapy
histone deacetylase inhibitor TS treatment: ↑ miR-16-5p
∆ HDAC3: ↑ miR-16-5p
[21]
TLN1
T98G and A172
TIIA treatment: ↑ miR-16-5p
↑↑ miR-16-5p: ↓ proliferation, migration, invasion
[22]
Neuroendocrine tumors
SSTR2
INS-1 rat insulinoma cell line and GH3 rat pituitary GH- and PRL-producing cell line
octreotide treatment: ↑ miR-16-5p
↑↑ miR-16-5p: ↓
Proliferation, ↑ SSTR2 expression
[26]
Chordoma
Smad3
U-CH1 and U-CH2
↑↑ miR-16-5p: ↓ proliferation, migration, invasion
[27]
LINC00662, RNF144B
U-CH1 and U-CH2
∆ LINC00662 (which sponges miR-16-5p): ↓ proliferation, migration, invasion, colony formation, and EMT process
[28]
Gastric cancer
PD-L1
PBMCs and CD3+ T cells
AGS and NCI-N87
M1 Macrophage-Secreted Exosomes Carrying miR-16-5p: ↑ polarization of macrophages to its M1 phenotype, and T cell activation, ↓ PD-L1 expression
[29]
Smad3
BSG823 and SGC-7901
Melatonin treatment: ↑ miR-16-5p: ↓ proliferation, ↑ apoptosis
[30]
LINC00649, YAP1, Hippo signaling pathway
MGC-803 and SGC-7901
∆ LINC00649 (which sponges miR-16-5p): ↓ proliferation, migration, viability, ↑ apoptosis
[31]
LINC00473, CCND2
BGC823, AGS, MKN-45, NCI-N87, GES-1
∆ LINC00473 (which sponges miR-16-5p): ↓ proliferation, migration, invasion, ↑ apoptosis, cell arrest
[32]
Lung cancer
WEE1
GLC-82 and HTB-182
Quercetin: ↑ miR-16-5p
↑↑ miR-16-5p: ↓ proliferation, colony formation, viability, ↑ apoptosis, and radiosensitivity
[33]
Linc00210, PTK2
BEAS-2B, A549, Calu-3, H1299, SPCA-1, and PC-9
∆ Linc00210 (which sponges miR-16-5p): ↓ proliferation, invasion, ↑ apoptosis
[34]
XIST, WEE1
H157, HCC827, A549 and H838
∆ XIST (which sponges miR-16-5p): ↓ colony formation, viability, ↑ apoptosis, and radiosensitivity
[35]
Colorectal cancer
PVT1, VEGFA, VEGFR1, AKT
signaling
FHC, HCT116 and SW480, and HEK293T
∆ PVT1 (which sponges miR-16-5p): ↓ proliferation, migration, and invasion
↑↑ miR-16-5p: ↓ proliferation, migration, and invasion
[36]
 
ALDH1A3, PKM2
HCT116, LoVo, DLD1 and RKO
CuET treatment: ↑ miR-16-5p and miR-15b-5p: ↓ glycolysis, viability and ↑ G2/M-phase arrest and apoptosis
∆ ALDH1A3 (a target of mir-16-5p): ↓ viability and clonogenicity
[37]
ITGA2
Caco-2, SW480,
SW620, LoVo, and HT29
↑↑ miR-16-5p: ↓ proliferation, migration, and invasion, ↑ apoptosis
∆ miR-16-5p: ↑ proliferation, migration, and invasion, ↓ apoptosis
[38]
Erythroleukemia
 
MEL cells
↑↑ miR-16-5p: ↑ erythroid differentiation of MEL cells by regulating ribosome biogenesis
[39]
Prostate cancer
AKT3
 
↑↑ miR-16-5p: ↓ cell survival, ↑ cell cycle distribution and apoptosis
[40]
Cyclin D1/E1-pRb-E2F1 pathway
LNCaP
Ionizing radiation: ↑ miR-16-5p: ↓ proliferation, viability, and ↑ G0/G1 phase arrest
[41]
Chondrosarcomas
VEGF-A, PI3K/Akt signaling
JJ012, SW1353
Resistin treatment: ↓ miR-16-5p: ↑ VEGF-A-dependent EPCs angiogenesis
[42]
Giant cell tumor of bone
 
BMM cells
↑↑ miR-16-5p: ↓ RANKL-induced osteoclastogenesis
[43]
Papillary thyroid carcinoma
SNHG12
PTC cell lines
∆ SNHG12 (which sponges miR-16-5p): ↓ proliferation, migration and invasion and ↑ apoptosis
[44]
Renal cell carcinoma
PVT1
HK-2, A498, 786-O, ACHN and Caki-1
∆ PVT1 (which sponges miR-16-5p): ↓ proliferation, migration invasion, EMT process, and ↑ apoptosis
∆ miR-16-5p: ↑ proliferation, migration invasion, EMT process, and ↓ apoptosis
[45]
Bladder cancer
BIMP1/NFκB signaling pathway
T24 and 5637
↑↑ miR-16-5p: ↓ viability, ↑ autophagy and apoptosis
[46]
LINC00649, JARID2
HCV-29, UMUC2, SW780, and T24
∆ LINC00649 (which sponges miR-16-5p): ↓ proliferation, migration, and invasion
[47]
Cholangiocarcinoma
R-2HG, ERα, YAP1
QBC939, HuCCT1, and HEK293T
IDH mutations: ↑ R-2HG production
↑ R-2HG: ↑ degradation of FTO so ↓ protein translation of the ERα: ↑ miR-16-5p: ↓ YAP1: ↓ proliferation and cell growth
[48]

Animal studies

The tumor suppressor role of miR-16-5p has been verified in animal models of different types of cancers. Studies in these models have shown that over-expression of this miRNA or modulation of expression of lncRNAs that sponge this miRNA can block carcinogenic processes. For instance, transplantation of miR-15a‐, miR‐15b‐ and miR‐16‐expressing neuroblastoma cells into extremely immunodeficient mice has suppressed formation of tumors as well as expression of MYCN, suggesting that these miRNAs have a tumor suppressor role in neuroblastoma through targeting MYCN [7]. Another study in xenograft model of neuroblastoma has shown that knock down of the miR-16-5p-targeting circ-CUX1 leads to reduction of tumor growth [8].
In animal models of hepatocellular carcinoma, up-regulation of AGAP2-AS1 has enhanced tumor growth via down-regulating miR-16-5p [11]. Moreover, down-regulation of TTN-AS1 decreases tumor size and resistance to sorafenib through enhancement of expression of miR-16-5p [13]. In cervical cancer models, silencing of miR-16-5p target, PDK4 has enhanced efficacy of chemotherapy [16]. Moreover, silencing of DLX6-AS1 which targets miR-16-5p decreases tumor size [25]. Other studies in breast cancer, chordoma/chondrosarcoma, gastric cancer, lung cancer, colorectal cancer, bladder cancer and cholangiocarcinoma have confirmed a tumor suppressor role for miR-16-5p (Table 2).
Table 2
Function of miR-16-5p or its partners in animal models (∆: knock-down or deletion)
Tumor Type
Animal models
Results
Reference
Neuroblastoma
NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice
↑↑ miR-16-5p: ↓ bioluminescence, tumor size, and tumor weight
[7]
BALB/c nude mice
∆ Circ-CUX1 (which suppresses miR-16-5p): ↓ tumor size, tumor weight, and tumor growth
[8]
Hepatocellular carcinoma
female BALB/c nude mice
↑↑ AGAP2-AS1: ↑ tumor growth and metastasis
∆ AGAP2-AS1: ↓ tumor growth and metastasis
[11]
male Balb/c nude mice
∆ TTN-AS1 (which suppresses miR-16-5p): ↓ tumor size, tumor weight, sorafenib resistance
[13]
male BALB/c nude mice
∆ SNHG22 (which suppresses the transcription of miR-16-5p): ↓ tumor growth and angiogenesis
[14]
Cervical cancer
BALB/c nude mice
∆ PDK4 (a target of miR-16-5p): ↑ chemotherapy efficiency
[16]
BALB/c nude mice
∆ DLX6-AS1 (which sponges miR-16-5p): ↓ tumor sizes, volumes, and weights
[25]
Breast cancer
BALB/c nude mice
∆ mir-16-5p: ↑ tumor volume, proliferation and metastasis
[17]
nude mice
↑↑ mir-16-5p: ↓ tumor growth
[19]
BALB/c nude mice
∆ ATXN8OS (which sponges miR-16-5p): ↑ tamoxifen sensitivity
[20]
Chordoma
BALB/c athymic nude mice
↑↑ mir-16-5p: ↓ tumor volume and proliferation
[27]
BALB/c nude mice
∆ LINC00662 (which sponges miR-16-5p): ↓ tumor volumes and tumor weight
[28]
Gastric cancer
BALB/c mice and NOD/SCID nude mice
M1 macrophage-secreted exosomes carrying miR-16-5p: ↓ tumor growth, volume and weight
[29]
female BALB/c nude mice
∆ LINC00649 (which sponges miR-16-5p): ↓ tumor growth
[31]
 
female BALB/c-nude mice
∆ LINC00649 (which sponges miR-16-5p): ↓ tumor growth, tumor weight proliferation, and metastasis
[32]
Lung cancer
nude mice
∆ LINC00649 (which sponges miR-16-5p): ↓ tumor growth, volume and weight
[34]
Colorectal cancer
male BALB/c nude mice
↑↑ mir-16-5p: ↓ tumor volume and weight
[36]
male nude mice
CuET treatment: ↓ tumor volume and growth, ↑ apoptosis
[37]
BALB/c nude mice
↑↑ mir-16-5p: ↓ tumor volume and growth
[38]
Chondrosarcoma
male nude mice
↑↑ Resistin: vessel markers VEGF-A and CD31, EPC markers CD34 and CD133, and vessel formation
[42]
Bladder cancer
male BALB/c nude mice
↑↑ miR-16-5p: ↓ tumor volume, weight, and growth
[46]
Cholangiocarcinoma
female nude mice
↑↑ R-2HG (which increases levels of miR-16-5p) : ↓ tumor growth
[48]

Human studies

Down-regulation of miR-16-5p has been verified in clinical samples obtained from patients with different malignancies. Moreover, AGAP2-AS1 that decreases miR-16-5p levels has been shown to be up-regulated in hepatocellular carcinoma tissues, particularly in metastatic and recurrent ones. In addition, expression levels of AGAP2-AS1 and miR-16-5p have been correlated with clinical parameters and poor prognosis of patients with this type of cancer [11]. In neuroblastoma, up-regulation in circ-CUX1 that sponges miR-16-5p has been correlated with advanced TNM stage, low differentiation grade and lymph node metastasis [8]. In breast cancer patients, miR-16-5p has been shown to have low expression. Notably, patients with low expression of miR-16-5p have been found to have a lower survival rate compared with those having high expression of miR-16-5p [17].
In the majority of CLL cases, miR-15a and miR-16-1 have been shown to be lost or down-regulated [6]. Moreover, assessment of GO database has led to identification of enrichment of MCL1 Apoptosis Regulator, BCL2 Family Member (MCL1), B-cell lymphoma 2 (BCL2), ETS Proto-Oncogene 1 (ETS1), or Jun Proto-Oncogene, AP-1 Transcription Factor Subunit (JUN) in miR-16 signature. Notably, these genes are involved in the regulation of apoptosis and cell cycle [6].
Several studies have reported down-regulation of this miRNA in nearly all examined malignant tissues except for ovarian cancer tissues. Similarly, lncRNAs or circRNAs that decrease expression of miR-16-5p have been found to be up-regulated in cancer samples compared with non-cancerous controls (Table 3).
Table 3
Dysregulation of miR-16-5p or its partners in clinical samples (NB: Neuroblastoma, FAM: fetal adrenal medulla, ANCTs: adjacent non-cancerous tissues, OS: Overall survival, TNM: tumor-node‐metastasis, ccRCC: clear cell renal cell carcinoma)
Tumor type
samples
Expression of miR-16-5p or other genes
(Tumor vs. Normal)
Kaplan-Meier analysis (impact of miR-16-5p dysregulation)
Association of expression of miR-16-5p or expression of other genes with clinicopathologic characteristics
Method by which RNA was detected
Reference
Neuroblastoma
R2 database, containing 105 NB patients
Down
   
[7]
50 pairs of tumor tissues and FAM tissues
Upregulation in circ-CUX1 (which sponges miR-16-5p)
Lower OS
Upregulation in circ-CUX1 was correlated with advanced TNM stage, low differentiation grade and lymph node metastasis.
 
[8]
Osteosarcoma
40 pairs of tumor tissues and ANCTs
Down
Lower OS
 
SYBR® Premix Ex TaqTM Kit
[9]
51 pairs of tumor tissues and ANCTs
Upregulation in LINC00662 (which sponges miR-16-5p)
Lower OS
Upregulation in LINC00662 was correlated with distant metastasis, TNM stage, and tumor size.
 
[23]
30 pairs of tumor tissues and ANCTs
Upregulation in hsa_circ_0005721 (which sponges miR-16-5p)
  
SYBR®Premix Ex Taq™
[24]
Hepatocellular carcinoma
137 pairs of tumor tissues and ANCTs
Upregulation in AGAP2-AS1 (which sponges miR-16-5p)
Downregulation in miR-16-5p
 
Upregulation in LINC00662 was correlated with large tumor size, metastasis, recurrence and high histological grade tissues
-
[11]
100 pairs of tumor tissues and ANCTs
Downregulation in miR-16-5p
  
SYBR PrimeScriptTM RT-PCR Kit
[12]
60 pairs of tumor tissues and ANCTs
Upregulation in SNHG22
(which suppresses transcription of miR-16-5p)
Lower OS
 
SYBR Green PCR kit
[14]
Cervical cancer
63 pairs of tumor tissues and ANCTs
Upregulation in CARM1 (a target of miR-16-5p)
Downregulation in miR-16-5p
 
Upregulation in CARM1 was correlated with higher clinical
staging and poorer tumor differentiation
SYBR Green PCR kit
[15]
Gliomas
72 pairs of tumor tissues and ANCTs
Downregulation in miR-16-5p
  
SYBR Green I fluorescence method
[17]
GEO and TCGA databases
Upregulation in ANLN (a target of miR-16-5p)
Lower OS
  
[18]
40 pairs of tumor tissues and ANCTs
Downregulation in miR-16-5p
  
SYBR Green kit
[19]
22 pairs of tumor tissues and ANCTs
Upregulation in ATXN8OS
(which sponges miR-16-5p)
  
SYBR® Premix Ex TaqTM reagent
[20]
Gliomas
79 patients with astrocytic gliomas and 9 non-neoplastic brain samples
Downregulation in miR-16-5p
  
TaqMan probe
[21]
Chordoma
12 chordoma tissues and 12 nucleus pulposus tissues
10 chordoma tissues and 5 nucleus pulposus tissues
Downregulation in miR-16-5p
  
SYBR-Green PCR Master Mix
[27]
30 pairs of tumor tissues and ANCTs
Upregulation in LINC00662
(which sponges miR-16-5p)
  
RT2 SYBR Green FAST Mastermix or miScript SYBR Green PCR Kit
[28]
Gastric cancer
54 pairs of tumor tissues and ANCTs
TCGA dataset
Downregulation in miR-16-5p
  
One-Step TB Green TM PrimeScript
TM RT-PCR kit
[31]
53 pairs of tumor tissues and ANCTs
Upregulation in Linc00210 (which sponges miR-16-5p)
 
Upregulation in LINC00473 was correlated with a higher risk of lymphatic metastasis, a higher incidence of vascular cancer embolus, and advanced TNM stage.
TB Premix Ex Taq
[32]
Lung cancer
40 pairs of tumor tissues and ANCTs
Upregulation in Linc00210 (which sponges miR-16-5p)
  
SYBR Premix Ex Taq II and Perfect Real Time
[34]
31 pairs of tumor tissues and ANCTs
Upregulation in XIST (which sponges miR-16-5p)
  
SYBR Green Master Mix
[35]
Colorectal cancer
72 pairs of tumor tissues and ANCTs
Upregulation in PVT1 (which sponges miR-16-5p)
Lower OS
Upregulation in PVT1 was significantly correlated with lymph node metastasis, distant metastasis, and TNM (tumor, node, metastasis) stage
SYBR Green
[36]
42 pairs of tumor tissues and ANCTs
Upregulation in ALDH1A3 (a target of miR-16-5p)
Lower OS
  
[37]
GEO database: GSE75970, GSE74602, GSE89076,
and GSE10950
Upregulation in ITGA2 (a target of miR-16-5p)
   
[38]
Chronic lymphocytic leukemia
224 CLL cases and 224 matched controls
miR-16-5p levels were unrelated to CLL risk.
  
TaqMan
probes
[49]
Chondrosarcoma
9 human chondrosarcoma tissues and 9 normal cartilage
Downregulation in miR-16-5p
   
[42]
Giant cell tumor of bone
17 GCT tissue and 4 cancellous bone as controls
Downregulation in miR-16-5p
  
iTaq™ Universal SYBR Green Supermix
[43]
Ovarian cancer
142 ovarian cancer patients, and 97 healthy controls
Upregulation in miR-16-5p
No correlation between the gene expression levels, and the survival
time
  
[50]
Renal cell carcinoma
25 patients with ccRCC
Upregulation in PVT1 (which sponges miR-16-5p)
 
Upregulation in PVT1 was correlated with TNM stage, Fuhrman grade, lymph node metastasis and tumor size.
SYBR Green
[45]

Discussion

miR-16-5p is an example of miRNAs with tumor suppressor role in almost all assessed tissues. This speculation is based on the observed down-regulation of this miRNA in nearly all examined malignant tissues except for ovarian cancer tissues. Moreover, a number of studies have reported up-regulation of lncRNAs that target this miRNA or specific targets of this miRNA. This miRNA has been found to be sponged by some lncRNAs and circRNAs, namely LINC00662, LINC00649, LINC00473, LINC00210, PVT1, XIST, AGAP2-AS1, DLX6-AS1, TTN-AS1, circ-CUX1 and hsa_circ_0005721. These observations indicate the complexity of the network through which miR-16-5p exerts its tumor suppressor effects. Moreover, abnormal up-regulation of the mentioned lncRNAs and circRNAs is regarded as a possible mechanism for down-regulation of miR-16-5p along with genomic variations in the genetic locus of this miRNA.
Phosphoinositide 3-kinase (PI3K)/AKT, Phosphatase and tensin homolog (PTEN)/AKT, NF-κB, Hippo and E1-pRb-E2F1 pathways are among signaling pathways being affected by dysregulation of miR-16-5p. Thus, down-regulation of miR-16-5p can lead to over-activity of cancer-related signals enhancing cell survival.
Down-regulation of miR-16-5p or up-regulation of lncRNAs/circRNAs that sponge this miRNA has been shown to be associated with malignant features of different cancers such as neuroblastoma, osteosarcoma, renal cell carcinoma and colorectal cancer, indicating a role for miR-16-5p as a prognostic marker in human cancers. In fact, down-regulation of this miRNA has been detected in samples with low level of differentiation and high propensity to local and distant metastases. Thus, patient with low levels of expression of this miRNA has exhibited poor clinical outcomes.
Since this miRNA can be detected in the peripheral blood, it represents a novel non-invasive strategy for early detection of cancer. However, since it is down-regulated in several types of cancers, the type of cancer cannot be detected through this route. Moreover, evaluation of levels of miR-16-5p in cancer patients can be used for follow-up after removal of primary tumor.
The mechanism behind down-regulation of miR-16-5p in malignant tissues is not investigated thoroughly, although deletion in the genomic region coding this miRNA is a putative mechanism. Moreover, up-regulation of lncRNAs/circRNAs that sponge this miRNA is a well-established mechanism for its down-regulation in different cancers. Induction of DNA methylation in miR-16-5p is another mechanism of down-regulation of this miRNA in cancers [14]. Future studies are needed to find possible epigenetic alterations that affect transcription of precursor of miR-16-5p.
Different studies have shown the effects of miR-16-5p in regulation of chemosensitivity, radiosensitivity as well as response to the targeted therapy by sorafenib. From a clinical point of view, up-regulation of miR-16-5p is a potentially effective modality for suppression of tumor growth and defeating chemotherapy resistance. However, introduction of miR-16-5p mimic into cancerous cells needs a specific strategy to shield the miRNA mimics from self-hydrolysis or degradation by RNases. Without these considerations, the short half-life of naked RNA mimics reduces the potential effects of miRNAs [51]. Moreover, issues regarding the toxicity or nonspecific cell-targeting nature of miRNA carriers should be solved. These issues have attenuated the pace of entering miRNA mimics into the clinical setting.
Cumulatively, miR-16-5p is a putative tumor suppressor miRNA that can be used as a therapeutic modality in different cancers. However, the biosafety and bioavailability issues should be solved before introduction of this modality in clinical settings.

Acknowledgements

The authors would like to thank the clinical Research Development Unit (CRDU) of Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran for their support, cooperation and assistance throughout the period of study

Declarations

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. Informed consent forms were obtained from all study participants. The study protocol was approved by the ethical committee of Shahid Beheshti University of Medical Sciences. All methods were performed in accordance with the relevant guidelines and regulations.
Consent of publication.
Not applicable.

Competing interests

The authors declare they have no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Wahid F, Shehzad A, Khan T, Kim YY. MicroRNAs: synthesis, mechanism, function, and recent clinical trials. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research. 2010;1803(11):1231–43. Wahid F, Shehzad A, Khan T, Kim YY. MicroRNAs: synthesis, mechanism, function, and recent clinical trials. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research. 2010;1803(11):1231–43.
2.
Zurück zum Zitat Peng Y, Croce CM. The role of MicroRNAs in human cancer. Signal Transduct Target therapy. 2016;1(1):1–9.CrossRef Peng Y, Croce CM. The role of MicroRNAs in human cancer. Signal Transduct Target therapy. 2016;1(1):1–9.CrossRef
3.
Zurück zum Zitat Galvão-Lima LJ, Morais AH, Valentim RA, Barreto EJ. miRNAs as biomarkers for early cancer detection and their application in the development of new diagnostic tools. Biomed Eng Online. 2021;20(1):1–20.CrossRef Galvão-Lima LJ, Morais AH, Valentim RA, Barreto EJ. miRNAs as biomarkers for early cancer detection and their application in the development of new diagnostic tools. Biomed Eng Online. 2021;20(1):1–20.CrossRef
4.
Zurück zum Zitat Filipów S, Łaczmański Ł. Blood Circulating miRNAs as Cancer Biomarkers for Diagnosis and Surgical Treatment Response. Front Genet. 2019;10:169-. PubMed PMID: 30915102. eng.PubMedPubMedCentralCrossRef Filipów S, Łaczmański Ł. Blood Circulating miRNAs as Cancer Biomarkers for Diagnosis and Surgical Treatment Response. Front Genet. 2019;10:169-. PubMed PMID: 30915102. eng.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proceedings of the national academy of sciences. 2002;99(24):15524-9. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proceedings of the national academy of sciences. 2002;99(24):15524-9.
6.
Zurück zum Zitat Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M, et al. MiR-15a and miR-16-1 cluster functions in human leukemia. Proceedings of the National Academy of Sciences. 2008;105(13):5166-71. Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M, et al. MiR-15a and miR-16-1 cluster functions in human leukemia. Proceedings of the National Academy of Sciences. 2008;105(13):5166-71.
7.
Zurück zum Zitat Chava S, Reynolds CP, Pathania AS, Gorantla S, Poluektova LY, Coulter DW, et al. miR-15a‐5p, miR‐15b‐5p, and miR‐16‐5p inhibit tumor progression by directly targeting MYCN in neuroblastoma. Mol Oncol. 2020;14(1):180–96.PubMedCrossRef Chava S, Reynolds CP, Pathania AS, Gorantla S, Poluektova LY, Coulter DW, et al. miR-15a‐5p, miR‐15b‐5p, and miR‐16‐5p inhibit tumor progression by directly targeting MYCN in neuroblastoma. Mol Oncol. 2020;14(1):180–96.PubMedCrossRef
8.
Zurück zum Zitat Zhang X, Zhang J, Liu Q, Zhao Y, Zhang W, Yang H. Circ-CUX1 accelerates the progression of neuroblastoma via miR-16-5p/DMRT2 axis. Neurochem Res. 2020;45(12):2840–55.PubMedCrossRef Zhang X, Zhang J, Liu Q, Zhao Y, Zhang W, Yang H. Circ-CUX1 accelerates the progression of neuroblastoma via miR-16-5p/DMRT2 axis. Neurochem Res. 2020;45(12):2840–55.PubMedCrossRef
9.
Zurück zum Zitat Gu Z, Li Z, Xu R, Zhu X, Hu R, Xue Y, et al. miR-16-5p Suppresses Progression and Invasion of Osteosarcoma via Targeting at Smad3. Front Pharmacol. 2020;11:1324.PubMedPubMedCentralCrossRef Gu Z, Li Z, Xu R, Zhu X, Hu R, Xue Y, et al. miR-16-5p Suppresses Progression and Invasion of Osteosarcoma via Targeting at Smad3. Front Pharmacol. 2020;11:1324.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Yu J, Zhang H, Yan L, Chang F, Jia Z, Yang X. microRNA-16-5p targeted tetraspanin 15 gene to inhibit the proliferation, migration and invasion of osteosarcoma cell through phospoinositide 3-kinase/protein kinase B signaling pathway. Zhonghua yi xue za zhi. 2020;100(21):1668–75.PubMed Yu J, Zhang H, Yan L, Chang F, Jia Z, Yang X. microRNA-16-5p targeted tetraspanin 15 gene to inhibit the proliferation, migration and invasion of osteosarcoma cell through phospoinositide 3-kinase/protein kinase B signaling pathway. Zhonghua yi xue za zhi. 2020;100(21):1668–75.PubMed
11.
Zurück zum Zitat Liu Z, Wang Y, Wang L, Yao B, Sun L, Liu R, et al. Long non-coding RNA AGAP2-AS1, functioning as a competitive endogenous RNA, upregulates ANXA11 expression by sponging miR-16-5p and promotes proliferation and metastasis in hepatocellular carcinoma. J Experimental Clin Cancer Res. 2019;38(1):1–15.CrossRef Liu Z, Wang Y, Wang L, Yao B, Sun L, Liu R, et al. Long non-coding RNA AGAP2-AS1, functioning as a competitive endogenous RNA, upregulates ANXA11 expression by sponging miR-16-5p and promotes proliferation and metastasis in hepatocellular carcinoma. J Experimental Clin Cancer Res. 2019;38(1):1–15.CrossRef
12.
Zurück zum Zitat Cheng B, Ding F, Huang C, Xiao H, Fei F, Li J. Role of miR-16-5p in the proliferation and metastasis of hepatocellular carcinoma. Eur Rev Med Pharmacol Sci. 2019;23(1):137–45.PubMed Cheng B, Ding F, Huang C, Xiao H, Fei F, Li J. Role of miR-16-5p in the proliferation and metastasis of hepatocellular carcinoma. Eur Rev Med Pharmacol Sci. 2019;23(1):137–45.PubMed
13.
Zurück zum Zitat Zhou Y, Huang Y, Dai T, Hua Z, Xu J, Lin Y, et al. LncRNA TTN-AS1 intensifies sorafenib resistance in hepatocellular carcinoma by sponging miR-16-5p and upregulation of cyclin E1. Biomed Pharmacother. 2021;133:111030.PubMedCrossRef Zhou Y, Huang Y, Dai T, Hua Z, Xu J, Lin Y, et al. LncRNA TTN-AS1 intensifies sorafenib resistance in hepatocellular carcinoma by sponging miR-16-5p and upregulation of cyclin E1. Biomed Pharmacother. 2021;133:111030.PubMedCrossRef
14.
Zurück zum Zitat Zhang Y, Lu C, Cui H. Long non-coding RNA SNHG22 facilitates hepatocellular carcinoma tumorigenesis and angiogenesis via DNA methylation of microRNA miR-16-5p. Bioengineered. 2021;12(1):7446–58.PubMedPubMedCentralCrossRef Zhang Y, Lu C, Cui H. Long non-coding RNA SNHG22 facilitates hepatocellular carcinoma tumorigenesis and angiogenesis via DNA methylation of microRNA miR-16-5p. Bioengineered. 2021;12(1):7446–58.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Zhang S, Wang W, Wu X, Liu W, Ding F. miR-16‐5p modulates the radiosensitivity of cervical cancer cells via regulating coactivator‐associated arginine methyltransferase 1. Pathol Int. 2020;70(1):12–20.PubMedCrossRef Zhang S, Wang W, Wu X, Liu W, Ding F. miR-16‐5p modulates the radiosensitivity of cervical cancer cells via regulating coactivator‐associated arginine methyltransferase 1. Pathol Int. 2020;70(1):12–20.PubMedCrossRef
16.
Zurück zum Zitat Zhao Z, Ji M, Wang Q, He N, Li Y. miR-16-5p/PDK4-mediated metabolic reprogramming is involved in chemoresistance of cervical cancer. Mol Therapy-Oncolytics. 2020;17:509–17.CrossRef Zhao Z, Ji M, Wang Q, He N, Li Y. miR-16-5p/PDK4-mediated metabolic reprogramming is involved in chemoresistance of cervical cancer. Mol Therapy-Oncolytics. 2020;17:509–17.CrossRef
18.
Zurück zum Zitat Wang Z, Hu S, Li X, Liu Z, Han D, Wang Y, et al. MiR-16-5p suppresses breast cancer proliferation by targeting ANLN. BMC Cancer. 2021;21(1):1–12.CrossRef Wang Z, Hu S, Li X, Liu Z, Han D, Wang Y, et al. MiR-16-5p suppresses breast cancer proliferation by targeting ANLN. BMC Cancer. 2021;21(1):1–12.CrossRef
19.
Zurück zum Zitat Qu Y, Liu H, Lv X, Liu Y, Wang X, Zhang M, et al. MicroRNA-16-5p overexpression suppresses proliferation and invasion as well as triggers apoptosis by targeting VEGFA expression in breast carcinoma. Oncotarget. 2017;8(42):72400.PubMedPubMedCentralCrossRef Qu Y, Liu H, Lv X, Liu Y, Wang X, Zhang M, et al. MicroRNA-16-5p overexpression suppresses proliferation and invasion as well as triggers apoptosis by targeting VEGFA expression in breast carcinoma. Oncotarget. 2017;8(42):72400.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Zhang H, Zhang J, Dong L, Ma R. LncRNA ATXN8OS enhances tamoxifen resistance in breast cancer. Open Med. 2021;16(1):68–80.CrossRef Zhang H, Zhang J, Dong L, Ma R. LncRNA ATXN8OS enhances tamoxifen resistance in breast cancer. Open Med. 2021;16(1):68–80.CrossRef
21.
Zurück zum Zitat Krell A, Wolter M, Stojcheva N, Hertler C, Liesenberg F, Zapatka M, et al. MiR-16‐5p is frequently down‐regulated in astrocytic gliomas and modulates glioma cell proliferation, apoptosis and response to cytotoxic therapy. Neuropathol Appl Neurobiol. 2019;45(5):441–58.PubMed Krell A, Wolter M, Stojcheva N, Hertler C, Liesenberg F, Zapatka M, et al. MiR-16‐5p is frequently down‐regulated in astrocytic gliomas and modulates glioma cell proliferation, apoptosis and response to cytotoxic therapy. Neuropathol Appl Neurobiol. 2019;45(5):441–58.PubMed
22.
Zurück zum Zitat You S, He X, Wang M, Mao L, Zhang L. Tanshinone IIA. Suppresses Glioma Cell Proliferation, Migration and Invasion Both in vitro and in vivo Partially Through miR-16-5p/Talin-1 (TLN1) Axis. Cancer Manage Res. 2020;12:11309.CrossRef You S, He X, Wang M, Mao L, Zhang L. Tanshinone IIA. Suppresses Glioma Cell Proliferation, Migration and Invasion Both in vitro and in vivo Partially Through miR-16-5p/Talin-1 (TLN1) Axis. Cancer Manage Res. 2020;12:11309.CrossRef
23.
Zurück zum Zitat Yu M, Lu W, Cao Z, Xuan T. LncRNA LINC00662 Exerts an Oncogenic Effect on Osteosarcoma by the miR-16-5p/ITPR1 Axis. Journal of oncology. 2021;2021. Yu M, Lu W, Cao Z, Xuan T. LncRNA LINC00662 Exerts an Oncogenic Effect on Osteosarcoma by the miR-16-5p/ITPR1 Axis. Journal of oncology. 2021;2021.
24.
Zurück zum Zitat Xu M, Sun X, Liu Y, Chang L, Te Wang H, Wang S. hsa_circ_0005721 triggers proliferation, migration and invasion of osteosarcoma by upregulating the linear transcript TEP1. J BU ON: Official J Balkan Union Oncol. 2021;26(4):1588–94. Xu M, Sun X, Liu Y, Chang L, Te Wang H, Wang S. hsa_circ_0005721 triggers proliferation, migration and invasion of osteosarcoma by upregulating the linear transcript TEP1. J BU ON: Official J Balkan Union Oncol. 2021;26(4):1588–94.
25.
Zurück zum Zitat Xie F, Xie G, Sun Q. Long noncoding RNA DLX6-AS1 promotes the progression in cervical cancer by targeting miR-16-5p/ARPP19 axis. Cancer Biother Radiopharm. 2020;35(2):129–36.PubMedCrossRef Xie F, Xie G, Sun Q. Long noncoding RNA DLX6-AS1 promotes the progression in cervical cancer by targeting miR-16-5p/ARPP19 axis. Cancer Biother Radiopharm. 2020;35(2):129–36.PubMedCrossRef
26.
Zurück zum Zitat Jo H, Park Y, Kim J, Kwon H, Kim T, Lee J, et al. Elevated miR-16-5p induces somatostatin receptor 2 expression in neuroendocrine tumor cells. PLoS ONE. 2020;15(10):e0240107.PubMedPubMedCentralCrossRef Jo H, Park Y, Kim J, Kwon H, Kim T, Lee J, et al. Elevated miR-16-5p induces somatostatin receptor 2 expression in neuroendocrine tumor cells. PLoS ONE. 2020;15(10):e0240107.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Zhang H, Yang K, Ren T, Huang Y, Tang X, Guo W. miR-16-5p inhibits chordoma cell proliferation, invasion and metastasis by targeting Smad3. Cell Death Dis. 2018;9(6):1–13.CrossRef Zhang H, Yang K, Ren T, Huang Y, Tang X, Guo W. miR-16-5p inhibits chordoma cell proliferation, invasion and metastasis by targeting Smad3. Cell Death Dis. 2018;9(6):1–13.CrossRef
28.
Zurück zum Zitat Wang C, Wang Y, Wang J, Guo X. LINC00662 triggers malignant progression of chordoma by the activation of RNF144B via targeting miR-16-5p. Eur Rev Med Pharmacol Sci. 2020;24(3):1007–22.PubMed Wang C, Wang Y, Wang J, Guo X. LINC00662 triggers malignant progression of chordoma by the activation of RNF144B via targeting miR-16-5p. Eur Rev Med Pharmacol Sci. 2020;24(3):1007–22.PubMed
29.
Zurück zum Zitat Li Z, Suo B, Long G, Gao Y, Song J, Zhang M, et al. Exosomal miRNA-16-5p derived from M1 macrophages enhances T cell-dependent immune response by regulating PD-L1 in gastric cancer. Front Cell Dev Biology. 2020;8:1362.CrossRef Li Z, Suo B, Long G, Gao Y, Song J, Zhang M, et al. Exosomal miRNA-16-5p derived from M1 macrophages enhances T cell-dependent immune response by regulating PD-L1 in gastric cancer. Front Cell Dev Biology. 2020;8:1362.CrossRef
30.
Zurück zum Zitat Zhu C, Huang Q, Zhu H. Melatonin inhibits the proliferation of gastric cancer cells through regulating the miR-16-5p-Smad3 pathway. DNA Cell Biol. 2018;37(3):244–52.PubMedCrossRef Zhu C, Huang Q, Zhu H. Melatonin inhibits the proliferation of gastric cancer cells through regulating the miR-16-5p-Smad3 pathway. DNA Cell Biol. 2018;37(3):244–52.PubMedCrossRef
31.
Zurück zum Zitat Wang H, Di X, Bi Y, Sun S, Wang T. Long non-coding RNA LINC00649 regulates YES-associated protein 1 (YAP1)/Hippo pathway to accelerate gastric cancer (GC) progression via sequestering miR-16-5p. Bioengineered. 2021;12(1):1791–802.PubMedPubMedCentralCrossRef Wang H, Di X, Bi Y, Sun S, Wang T. Long non-coding RNA LINC00649 regulates YES-associated protein 1 (YAP1)/Hippo pathway to accelerate gastric cancer (GC) progression via sequestering miR-16-5p. Bioengineered. 2021;12(1):1791–802.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Zhuo S, Sun M, Bai R, Lu D, Di S, Ma T, et al. Long intergenic non-coding RNA 00473 promotes proliferation and migration of gastric cancer via the miR-16-5p/CCND2 axis and by regulating AQP3. Cell Death Dis. 2021;12(5):1–14.CrossRef Zhuo S, Sun M, Bai R, Lu D, Di S, Ma T, et al. Long intergenic non-coding RNA 00473 promotes proliferation and migration of gastric cancer via the miR-16-5p/CCND2 axis and by regulating AQP3. Cell Death Dis. 2021;12(5):1–14.CrossRef
33.
Zurück zum Zitat Wang Q, Chen Y, Lu H, Wang H, Feng H, Xu J, et al. Quercetin radiosensitizes non-small cell lung cancer cells through the regulation of miR‐16‐5p/WEE1 axis. IUBMB Life. 2020;72(5):1012–22.PubMedCrossRef Wang Q, Chen Y, Lu H, Wang H, Feng H, Xu J, et al. Quercetin radiosensitizes non-small cell lung cancer cells through the regulation of miR‐16‐5p/WEE1 axis. IUBMB Life. 2020;72(5):1012–22.PubMedCrossRef
34.
Zurück zum Zitat Peng Q, Chen Y, Li C. Long noncoding RNA Linc00210 promotes non-small cell lung cancer progression via sponging miR-16-5p/PTK2 axis. Eur Rev Med Pharmacol Sci. 2020;24(18):9438–52.PubMed Peng Q, Chen Y, Li C. Long noncoding RNA Linc00210 promotes non-small cell lung cancer progression via sponging miR-16-5p/PTK2 axis. Eur Rev Med Pharmacol Sci. 2020;24(18):9438–52.PubMed
35.
Zurück zum Zitat Du R, Jiang F, Yin Y, Xu J, Li X, Hu L, et al. Knockdown of lncRNA X inactive specific transcript (XIST) radiosensitizes non-small cell lung cancer (NSCLC) cells through regulation of miR-16-5p/WEE1 G2 checkpoint kinase (WEE1) axis. Int J ImmunoPathol Pharmacol. 2021;35:2058738420966087.PubMedPubMedCentralCrossRef Du R, Jiang F, Yin Y, Xu J, Li X, Hu L, et al. Knockdown of lncRNA X inactive specific transcript (XIST) radiosensitizes non-small cell lung cancer (NSCLC) cells through regulation of miR-16-5p/WEE1 G2 checkpoint kinase (WEE1) axis. Int J ImmunoPathol Pharmacol. 2021;35:2058738420966087.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Wu H, Wei M, Jiang X, Tan J, Xu W, Fan X, et al. lncRNA PVT1 promotes tumorigenesis of colorectal cancer by stabilizing miR-16-5p and interacting with the VEGFA/VEGFR1/AKT axis. Mol Therapy-Nucleic Acids. 2020;20:438–50.CrossRef Wu H, Wei M, Jiang X, Tan J, Xu W, Fan X, et al. lncRNA PVT1 promotes tumorigenesis of colorectal cancer by stabilizing miR-16-5p and interacting with the VEGFA/VEGFR1/AKT axis. Mol Therapy-Nucleic Acids. 2020;20:438–50.CrossRef
37.
Zurück zum Zitat Huang X, Hou Y, Weng X, Pang W, Hou L, Liang Y, et al. Diethyldithiocarbamate-copper complex (CuET) inhibits colorectal cancer progression via miR-16-5p and 15b-5p/ALDH1A3/PKM2 axis-mediated aerobic glycolysis pathway. Oncogenesis. 2021;10(1):1–16.CrossRef Huang X, Hou Y, Weng X, Pang W, Hou L, Liang Y, et al. Diethyldithiocarbamate-copper complex (CuET) inhibits colorectal cancer progression via miR-16-5p and 15b-5p/ALDH1A3/PKM2 axis-mediated aerobic glycolysis pathway. Oncogenesis. 2021;10(1):1–16.CrossRef
38.
Zurück zum Zitat Xu Y, Shen L, Li F, Yang J, Wan X, Ouyang M. microRNA-16‐5p‐containing exosomes derived from bone marrow‐derived mesenchymal stem cells inhibit proliferation, migration, and invasion, while promoting apoptosis of colorectal cancer cells by downregulating ITGA2. J Cell Physiol. 2019;234(11):21380–94.PubMedCrossRef Xu Y, Shen L, Li F, Yang J, Wan X, Ouyang M. microRNA-16‐5p‐containing exosomes derived from bone marrow‐derived mesenchymal stem cells inhibit proliferation, migration, and invasion, while promoting apoptosis of colorectal cancer cells by downregulating ITGA2. J Cell Physiol. 2019;234(11):21380–94.PubMedCrossRef
39.
Zurück zum Zitat Papagiannopoulos CI, Theodoroula NF, Vizirianakis IS. miR-16-5p Promotes Erythroid Maturation of Erythroleukemia Cells by Regulating Ribosome Biogenesis. Pharmaceuticals. 2021;14(2):137.PubMedPubMedCentralCrossRef Papagiannopoulos CI, Theodoroula NF, Vizirianakis IS. miR-16-5p Promotes Erythroid Maturation of Erythroleukemia Cells by Regulating Ribosome Biogenesis. Pharmaceuticals. 2021;14(2):137.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Wang F, Wang W, Lu L, Xie Y, Yan J, Chen Y, et al. MicroRNA–16–5p regulates cell survival, cell cycle and apoptosis by targeting AKT3 in prostate cancer cells. Oncol Rep. 2020;44(3):1282–92.PubMedCrossRef Wang F, Wang W, Lu L, Xie Y, Yan J, Chen Y, et al. MicroRNA–16–5p regulates cell survival, cell cycle and apoptosis by targeting AKT3 in prostate cancer cells. Oncol Rep. 2020;44(3):1282–92.PubMedCrossRef
41.
Zurück zum Zitat Wang F, Mao A, Tang J, Zhang Q, Yan J, Wang Y, et al. microRNA-16‐5p enhances radiosensitivity through modulating Cyclin D1/E1–pRb–E2F1 pathway in prostate cancer cells. J Cell Physiol. 2019;234(8):13182–90.PubMedCrossRef Wang F, Mao A, Tang J, Zhang Q, Yan J, Wang Y, et al. microRNA-16‐5p enhances radiosensitivity through modulating Cyclin D1/E1–pRb–E2F1 pathway in prostate cancer cells. J Cell Physiol. 2019;234(8):13182–90.PubMedCrossRef
42.
Zurück zum Zitat Chen S-S, Tang C-H, Chie M-J, Tsai C-H, Fong Y-C, Lu Y-C, et al. Resistin facilitates VEGF-A-dependent angiogenesis by inhibiting miR-16-5p in human chondrosarcoma cells. Cell Death Dis. 2019;10(1):1–12.CrossRef Chen S-S, Tang C-H, Chie M-J, Tsai C-H, Fong Y-C, Lu Y-C, et al. Resistin facilitates VEGF-A-dependent angiogenesis by inhibiting miR-16-5p in human chondrosarcoma cells. Cell Death Dis. 2019;10(1):1–12.CrossRef
43.
Zurück zum Zitat Sang S, Zhang Z, Qin S, Li C, Dong Y. MicroRNA-16-5p inhibits osteoclastogenesis in giant cell tumor of bone. BioMed research international. 2017;2017. Sang S, Zhang Z, Qin S, Li C, Dong Y. MicroRNA-16-5p inhibits osteoclastogenesis in giant cell tumor of bone. BioMed research international. 2017;2017.
44.
Zurück zum Zitat Feng X, Dong X, Wu D, Zhao H, Xu C, Li H. Long noncoding RNA small nucleolar RNA host gene 12 promotes papillary thyroid carcinoma cell growth and invasion by targeting miR-16-5p. Histol Histopathol. 2019;35(2):217–24.PubMed Feng X, Dong X, Wu D, Zhao H, Xu C, Li H. Long noncoding RNA small nucleolar RNA host gene 12 promotes papillary thyroid carcinoma cell growth and invasion by targeting miR-16-5p. Histol Histopathol. 2019;35(2):217–24.PubMed
45.
Zurück zum Zitat Ren Y, Huang W, Weng G, Cui P, Liang H, Li Y. lncrna PVT1 promotes proliferation, invasion and epithelial–mesenchymal transition of renal cell carcinoma cells through downregulation of mir-16-5p. OncoTargets and therapy. 2019;12:2563.PubMedPubMedCentralCrossRef Ren Y, Huang W, Weng G, Cui P, Liang H, Li Y. lncrna PVT1 promotes proliferation, invasion and epithelial–mesenchymal transition of renal cell carcinoma cells through downregulation of mir-16-5p. OncoTargets and therapy. 2019;12:2563.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat He J, Qiu Z, Zhang H, Gao Z, Jiang Y, Li Z, et al. MicroRNA–16–5p/BIMP1/NF–κB axis regulates autophagy to exert a tumor–suppressive effect on bladder cancer. Mol Med Rep. 2021;24(2):1–10.CrossRef He J, Qiu Z, Zhang H, Gao Z, Jiang Y, Li Z, et al. MicroRNA–16–5p/BIMP1/NF–κB axis regulates autophagy to exert a tumor–suppressive effect on bladder cancer. Mol Med Rep. 2021;24(2):1–10.CrossRef
47.
Zurück zum Zitat Liu Y, Huang X, Guo L, Luo N. LINC00649 Facilitates the Cellular Process of Bladder Cancer Cells via Signaling Axis miR-16-5p/JARID2. Urologia Internationalis. 2021:1–9. Liu Y, Huang X, Guo L, Luo N. LINC00649 Facilitates the Cellular Process of Bladder Cancer Cells via Signaling Axis miR-16-5p/JARID2. Urologia Internationalis. 2021:1–9.
48.
Zurück zum Zitat Gao Y, Ouyang X, Zuo L, Xiao Y, Sun Y, Chang C, et al. R-2HG downregulates ERα to inhibit cholangiocarcinoma via the FTO/m6A-methylated ERα/miR16-5p/YAP1 signal pathway. Mol Therapy-Oncolytics. 2021;23:65–81.CrossRef Gao Y, Ouyang X, Zuo L, Xiao Y, Sun Y, Chang C, et al. R-2HG downregulates ERα to inhibit cholangiocarcinoma via the FTO/m6A-methylated ERα/miR16-5p/YAP1 signal pathway. Mol Therapy-Oncolytics. 2021;23:65–81.CrossRef
49.
Zurück zum Zitat Casabonne D, Benavente Y, Seifert J, Costas L, Armesto M, Arestin M, et al. Serum levels of hsa-miR‐16‐5p, hsa‐miR‐29a‐3p, hsa‐miR‐150‐5p, hsa‐miR‐155‐5p and hsa‐miR‐223‐3p and subsequent risk of chronic lymphocytic leukemia in the EPIC study. Int J Cancer. 2020;147(5):1315–24.PubMedCrossRef Casabonne D, Benavente Y, Seifert J, Costas L, Armesto M, Arestin M, et al. Serum levels of hsa-miR‐16‐5p, hsa‐miR‐29a‐3p, hsa‐miR‐150‐5p, hsa‐miR‐155‐5p and hsa‐miR‐223‐3p and subsequent risk of chronic lymphocytic leukemia in the EPIC study. Int J Cancer. 2020;147(5):1315–24.PubMedCrossRef
50.
Zurück zum Zitat Saral MA, Tuncer SB, Odemis DA, Erdogan OS, Erciyas SK, Saip P, et al. New biomarkers in peripheral blood of patients with ovarian cancer: high expression levels of miR-16-5p, miR-17-5p, and miR-638. Archives of Gynecology and Obstetrics. 2021:1–9. Saral MA, Tuncer SB, Odemis DA, Erdogan OS, Erciyas SK, Saip P, et al. New biomarkers in peripheral blood of patients with ovarian cancer: high expression levels of miR-16-5p, miR-17-5p, and miR-638. Archives of Gynecology and Obstetrics. 2021:1–9.
51.
Zurück zum Zitat Lee TJ, Yuan X, Kerr K, Yoo JY, Kim DH, Kaur B, et al. Strategies to Modulate MicroRNA Functions for the Treatment of Cancer or Organ Injury. Pharmacol Rev. 2020 Jul;72(3):639–67. PubMed PMID: 32554488. Pubmed Central PMCID: PMC7300323. Epub 2020/06/20. eng.PubMedPubMedCentralCrossRef Lee TJ, Yuan X, Kerr K, Yoo JY, Kim DH, Kaur B, et al. Strategies to Modulate MicroRNA Functions for the Treatment of Cancer or Organ Injury. Pharmacol Rev. 2020 Jul;72(3):639–67. PubMed PMID: 32554488. Pubmed Central PMCID: PMC7300323. Epub 2020/06/20. eng.PubMedPubMedCentralCrossRef
Metadaten
Titel
A review on the role of mir-16-5p in the carcinogenesis
verfasst von
Soudeh Ghafouri-Fard
Tayyebeh Khoshbakht
Bashdar Mahmud Hussen
Sara Tharwat Abdullah
Mohammad Taheri
Mohammad Samadian
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2022
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-022-02754-0

Weitere Artikel der Ausgabe 1/2022

Cancer Cell International 1/2022 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Update Onkologie

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