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

Open Access 01.12.2022 | Review

Functions and underlying mechanisms of miR-650 in human cancers

verfasst von: Yuanshuai Su, Qiuxian Zheng, Lingxiao Zhu, Xinyu Gu, Juan Lu, Lanjuan Li

Erschienen in: Cancer Cell International | Ausgabe 1/2022

Abstract

MicroRNAs (miRNAs) are one type of noncoding RNAs that interfere with mRNA translation to downregulate gene expression, which results in posttranscriptional gene silencing. Over the past two decades, miRNAs have been widely reported to impact the progression of malignant tumours by interfering with cancer initiation and progression; therefore, miRNAs represent potential new diagnostic and therapeutic tools. miR-650 is a newly identified miR, and increasing studies have demonstrated that miR-650 plays critical roles in cancer progression, such as mediating the Wnt signalling pathway/AXIN1 (axis inhibition protein 1) axis in hepatocellular carcinoma. Nevertheless, associations between the expression patterns and molecular mechanisms of miR-650 in cancer have not been comprehensively described. In this article, we review the existing evidence regarding the mechanisms by which miR-650 expression is altered and their relation to cancer. Moreover, the promising clinical application of miR-650 for diagnosis and treatment is highlighted.
Hinweise
Yuanshuai Su and Qiuxian Zheng contributed equally to this work

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
miRNAs
microRNAs
KRAS
Kirsten rat sarcoma viral oncogene homologue
PD-1
Programmed cell death protein 1
EC
Endometrial cancer
EMT
Epithelial to mesenchymal transition
ING4
Inhibitor of growth 4
HCC
Hepatocellular carcinoma
NASH
Non-alcoholic steatohepatitis
LC
Lung cancer
NSCLC
Non-small-cell lung cancer
LAD
Lung adenocarcinoma
CRC
Colorectal cancer
GC
Gastric cancer
AML
Acute myeloid leukaemia
EPOR
Erythropoietin receptor
CLL
Chronic lymphocyte leukaemia
TTFT
Time to first treatment
ATC
Anaplastic thyroid carcinoma
NF
Nuclear factor
SOCS3
Suppressor of cytokine signalling 3
LCCs
Large-cell carcinomas
LCSCs
Lung cancer stem cells
AXIN1
Axis inhibition protein 1
CDKN1B
Cyclin-dependent inhibitor 1B
DTIC
Dacarbazine
MGMT
Methylguanine-DNA-methyltransferase
TGF-β
Transforming growth factor-β
PPP2CA
Protein Phosphatase 2 Catalytic Subunit Alpha
CSR1
Cellular stress response
LATS2
Large tumor suppressor kinase 2 gene
JAK
Janus kinase
STAT3
Signal transducer and activator of transcription 3
MEG3
Maternally expressed gene 3
SLC34A2
Solute carrier family 34 member 2
ANXA2
Annexin 2
NDRG2
N-myc downstream regulated gene 2
ERK
Extracellular regulated protein kinases
RERG
Estrogen-regulated growth inhibitor
PHLPP2
pH domain and leucine rich repeat protein phosphatase 2
MAPK
Mitogen-activated protein kinases
GSK3β
Glycogen synthase kinase 3β
FAM83F
Family with sequence similarity 83member F
CDK1
Cyclin-dependent kinase

Background

Cancer is currently the major cause of death worldwide. By 2030, the number of cancer deaths is estimated to increase to 13 million [1], which will be an immense burden on the worldwide health system and especially on the health systems in low-income countries [2]. Primary cancer prevention, early detection and timely treatment are the most cost-effective to improve the prognosis and reduce the mortality of patients with cancer. Over the past few decades, great efforts have been made to reveal the molecular mechanisms of tumorigenesis, and its management has been dramatically revolutionized by accurately targeted therapies. For example, directly targeted drugs for KRAS (Kirsten rat sarcoma viral oncogene homologue), one of the most frequently mutated oncogenes, have obtained encouraging results in clinical trials [3]. Moreover, immune therapy aimed at immune checkpoints, such as inhibitors of programmed cell death protein 1 (PD-1), provides huge benefits to cancer patients [4]. Despite great improvements in diagnosis and treatment, the patient mortality associated with tumours are still a severe problem in both sexes. Hence, new molecular agents and strategies with high accuracy need to be identified and applied to the early diagnosis and effective treatment of human cancer.
As a class of endogenous and noncoding RNAs run the length of only 20–24 nt [5], miRNAs play a central role in cell proliferation and migration. By interacting with supplementary series in the 3′-untranslated regions of target mRNAs, miRNAs regulate target genes by repressing translation or degrading target mRNAs at the posttranscriptional level. In just over two decades, more than 2600 miRNAs have been uncovered, and the miR field has expanded considerably [6, 7]. Currently, increasing evidence from in vivo and in vitro experiments has revealed that miRNAs play crucial roles as multifunctional regulators of wide-ranging diseases, particularly in cancer. miRNAs are further categorized into tumour promoters and tumour suppressors [8]. For example, miR-34a, one of the best-investigated members of the miR-34 family, has been shown to play a tumour suppressive role in p53-mediated apoptosis by directly targeting the antiapoptotic protein sirtuin1 [9]. In head and neck cancer, miRNA-196a has been shown to promote cisplatin resistance by targeting (cyclin-dependent kinase inhibitor 1B (CDKN1B) [10]. Insights into the molecular mechanisms of miRNAs in cancer have made miRNAs attractive biomarkers for cancer diagnosis and novel therapeutic approaches.
In recent years, numerous studies have revealed that miR-650 plays vital roles in various tumours. miR-650 is up- or downregulated in various tumour types and can act as a tumour suppressor or oncogene. Moreover, miRNA mimics and molecules targeting miR-650 (anti-miR) have shown tremendous potential in preclinical development. One recent study reported that lncRNA MCTP1 upregulated the expression of miR-650 in endometrial cancer (EC) through the miR-650/SMAD7 axis [11] and then modulated the cell proliferation and invasion, and induced epithelial to mesenchymal transition (EMT) process of EC. Inhibitor of growth 4 (ING4) is a well-known tumour suppressor, and some reports have shown that the ING4 expression level is remarkably downregulated in tumour tissues by miR-650 [1218]. miR-650 also contributes to drug resistance, such as by upregulating dacarbazine (DTIC) resistance in melanoma through the lncRNA POU3F3/miR-650/MGMT (methylguanine-DNA-methyltransferase) axis [19], which turns miR-650 into a potential target for cancer treatment. In addition, miR-650 can influence other diseases, such as rheumatoid arthritis [20, 21], ulcerative colitis, and cerebral ischaemia/reperfusion injury. In this review, we systematically summarize the expression profiles and functions of miR-650 in various tumours, with special emphasis on its target genes, upstream regulators, and interacting molecules. Furthermore, we summarize the clinical application potential of targeting miR-650 in the oncology field.

Human studies of miR-650

Initially, miR-650 was identified by Cummins et al. [22] using a miRAGE cloning approach in colorectal cells. Over the next few years, it was quickly recognized that miR-650 is conserved and has extensive functional significance throughout the plant and animal kingdoms. Data from GeneCards (https://​www.​genecards.​org) reveal that miR-650 genes are located in the eukaryotic genome on chromosome 22. Sequence analysis and structural predictions in Das’s research [23] revealed that the miR-650 genes were emerged in multiple copies and overlapped with the leader exons of the immunoglobulin lambda variable 2–8 genes located on chromosome 22 in the same transcription orientation. Notably, the miR-650 gene is the only known miRNA gene that overlaps with immunoglobulin genes [24].

Expression of miR-650 in cancers

Hepatocellular carcinoma (HCC)

Hepatocellular carcinoma (HCC), the most frequent primary cancer, is the third major cause of tumor-related deaths [25]. It has the characteristics of a high degree of malignancy, poor treatment response and unfavourable prognosis [26] and represents a heavy public health burden. Multiple risk factors [2729] involved in the complex process of HCC have been widely reported, among which miR-650 is one of the key molecules. For the past few years, dysregulation of miR-650 in HCC has been widely reported. Han et al. [30] found that miR-650 expression significantly overexpressed in HCC tissues, especially in patients with tumour metastasis. Qin’s et al. [31] research revealed that the expression of Axin, which can inhibit the progression of HCC by targeting miR-650, was weak in HCC. Moreover, the overexpression of miR-650, which can be stimulated with benzo[a]pyrene, promotes the pathological process of fatty liver disease and HCC, as implicated in the work of Ge et al. [32]. Another bioinformatic analysis also demonstrated that the relative quantification of miR-650 in liver tissue was markedly increased in non-alcoholic steatohepatitis (NASH) groups [33]. In addition, elevated miR-650 expression was significantly associated with patient differentiation capability and advanced tumour TNM stage [34]. These findings combined with the above data based on cell lines and clinical samples indicate that miR-650 functions as a tumour promoter in HCC. As discussed above, the progression of HCC is closely related to the expression of miR-650, making it a promising biomarker and therapeutic target for the early prevention and applicable treatment of HCC. Nevertheless, the antitumour effect of miR-650 has rarely been reported in HCC.

miR-650 and lung cancer (LC)

Approximately 2.2 million cases of lung cancer (LC) were newly increased in 2020. LC causes the most cancer-induced deaths (18.0% of total cancer deaths) [35], of which non-small-cell lung cancer (NSCLC) accounts for the majority [36]. Accumulating studies have indicated that miR-650 acts as a tumour promoter in LC, including NSCLC and lung adenocarcinoma (LAD). In the present study, miR-650 was shown to highly express in NSCLC. Huang et al. [16] found that the expression level of miR-650 was substantially higher in LAD tissue samples than in adjacent normal controls. Moreover, the overexpression of miR-650 was dramatically correlated with the clinical characteristics of LAD patients, such as advanced tumour stage, high incidence of lymph node metastasis, and unfavourable prognosis [16]. Furthermore, downregulation of miR-650 reversed the docetaxel resistance of LAD cells [16]. Another study demonstrated that miR-650 overexpressed in NSCLC, and in vitro experiments indicated that miR-650 promoted the cancer cell proliferation and migration, which resulted in low overall survival date of NSCLC patients [13]. Moreover, a miR-650 inhibitor was shown to attenuate si-MEG3-induced promotion of the LC stem cell-like state, migration and invasion in NSCLC [37]. In spite of great advances in diagnostic and therapeutic methods, the overall survival rate of NSCLC patients is still below 15% [38]. Thus, more efforts are needed to explore new effective strategies for the diagnosis and clinical treatment of LC.

miR-650 and colorectal cancer (CRC)

According to Global Cancer Statistics, more than 1.9 million new cases of colorectal cancer (CRC) were estimated to occur in 2020, causing 935,000 deaths globally. Tumour metastasis develops in approximately 10% of patients in stage I or II and eventually leads to death within 5 years after exsection [39]. Currently, dysregulation of miR-650 has been researched in CRC, and miR-650 has been reported to act as both a tumour promoter or suppressor; therefore, the role and function of miR-650 in CRC remain controversial. Zhou et al. [39] discovered that the expression of miR-650 in CRC tissues positively correlated with the overall survival of patients. Furthermore, it repressed high-risk non-metastatic CRC progression [39] by inhibiting cell growth and invasion. In contrast, some reports have shown that miR-650 is upregulated and functions as an oncogene in CRC [14, 4042]. Based on what we already know, there is seemingly no consensus about the expression and function of miR-650 in CRC progression. Thus, more research is needed to comprehensively explore the roles of miR-650 in CRC.

miR-650 and gastric cancer (GC)

Gastric cancer (GC) was responsible for approximately 769,000 deaths in 2020, ranking fourth in mortality and fifth in incidence [43] globally. Due to the diagnosis of GC at early stages with complications, limited treatment options and poor prognoses, GC remains a great clinical challenge [44]. At present, the standard diagnostic methods for GC patients are gastroscopy and biopsy, but the utility of these methods is limited largely due to the invasiveness of GC and limited medical resources [45]. miR-650, as a new tumour biomarker in GC, has been investigated in recent years. One bioinformatic study based on 180 GC patients and 45 healthy individuals indicated that elevated miR-650 expression levels was significantly correlated with the existence of GC [45] and miRNA-650 are evaluated to be a promising and powerful non-invasive biomarker for the detection of GC. Previously, Zhang et al. [15] reported that overexpression of miR-650 promoted GC tumorigenesis in vivo and GC cell clonogenicity in vivo. Moreover, the overexpression of miR-650 has a positively association with the advancement of GC, as demonstrated in the work of Liu et al. [46] and An et al. [47].

miR-650 and glioma

Glioma, the most common primary tumour in the brain, accounts for approximately one-third of malignant cancers of the central nervous system [48]. Sun et al. [49] found that miR-650 expression was critically increased in glioma tissues and that was more frequently explored in tumours with a high WHO grade or low Karnofsky performance score (P < 0.001). Another study reported that miR-650 overexpressed in glioma tissues and cell lines. Furthermore, intensive expression was significantly correlated with the advanced tumour stage, lymph node metastasis and poor prognoses in glioma patients [50]. In contrast, Xu et al. [51] found that miR-650 was expressed at a low level in glioma tissues and in vitro cell lines.

miR-650 and melanoma

Cutaneous melanoma is a common tumour derived from the epidermis and mucosa [52]; it comprises 4-10% of all malignant cancers and is correlated with 75% of skin cancer-related deaths [53]. The death rates associated with melanoma have reportedly dropped rapidly after the introduction of new therapies, including targeted therapies for melanoma metastasis and immune checkpoint inhibitors [54, 55]. Liu et al. [56] recently found that the regulation of miR-650 has a negative correlation with melanoma progression. Moreover, further study showed that miR-650 overexpression alleviated MGMT-induced DTIC resistance in melanoma by increasing cell apoptosis [19], which indicated that miR-650 was a novel biomarker of great value for the evaluation of melanoma.

miR-650 and leukaemia

As the most frequent malignancy of the bone marrow, acute myeloid leukaemia (AML) has a high fatality rate [35]. Yuan et al. [57] revealed that miR-650 expression was reduced in AML, which contributed to leukaemia progression. Similarly, Gaine et al. [58] found that the expression of the erythropoietin receptor (EPOR) in t(12;21) B-ALL cells was higher than that in normal samples. Notably, EPOR expression is influenced by GATA2 and has an inverse correlation with miR-650 expression [58]. One study published in Blood reported that overexpression of miR-650 is correlated with a favourable chronic lymphocyte leukaemia (CLL) prognosis and affects the oncogenic capacity of B cells [24]. Furthermore, multivariate analysis showed that overexpression of miR-650 is an available independent predictor of survival and time to first treatment (TTFT) [24]. However, an inconsistent report that partly argued against the results above showed a significant increase in miR-650 expression in CLL patients [59]. Hence, the association between leukaemia and miR-650 needs further verification by expanding the number of patient samples included in the current work.

Other tumours

miR-650 has also been found to serve as an oncogene that is upregulated in various other cancers. In breast cancer, overexpression of miR-650 has been observed to induce the downregulation of the tumour suppressor ING4, which we previously mentioned is significantly correlated with EMT of breast cancer cells [60]. Another recent study revealed that the expression level of miR-650 is upregulated in EC, resulting in the progression of EC through the SMAD7-TGF-β (transforming growth factor-β) pathway [11]. In addition, miR-650 overexpressed in anaplastic thyroid carcinoma (ATC), where it promotes the proliferation and motility of cancer cells by targeting Protein Phosphatase 2 Catalytic Subunit Alpha (PPP2CA) [61]. In osteosarcoma, miR-650 has been reported to function as an oncogene. Yun et al. [18] revealed that overexpressing miR-650 decreased ING4 expression in human osteosarcoma cells and increased IL-6 transcription. Both ING-4 and IL-6 could modulate osteoblast and osteoclast differentiation. Moreover, miR-650 could upregulate the transcriptional activity of NF κB [18] and reduce the quantity of nuclear factor (NF) of κ light polypeptide gene enhancer. Interestingly, similarly significant upregulation of miR-650 has been observed in oral cancer, and the expression of miR-650 has been positively correlated with cancer cell proliferation, migration and invasion [62]. Likewise, Zuo et al. [63] reported that miR-650 could function as an onco-miR in prostate cancer by suppressing cellular stress response (CSR1) expression.
Overall, these findings have revealed that the profile of miR-650 expression depends on the type of cancer and that miR-650 is involved in the occurrence and development of various cancers. The overall expression files of miR-650 in various cancers and relative clinical features are presented in Tables 1 and 2. Even so, more studies are required to further explore the expression profiles of miR-650 in tumours.
Table 1
Expression of miR-650 in various cancer and relative clinical significance
Cancer type
Expression
Samples
Clinical significance
References
HCC
Upregulated
Tissues from 130 HCC patients
Microscopic vascular invasion, tumour volume, TNM stage
[30]
HCC
Upregulated
/
/
[31]
HCC
Upregulated
248 HCC tissues and 120 paracarcinomatous HCC tissues
Age, differentiation capability, tumour stage
[34]
FLD/HCC
Upregulated
/
Tumour metastasis
[32]
NSCLC
Upregulated
53 NSCLC tissues and adjacent normal lung tissues
/
[64]
NSCLC
Upregulated
49 NSCLC tissues and adjacent normal tissues
Overall survival rate
[13]
NSCLC
Upregulated
/
/
[37]
LAD
Upregulated
96 LAD tissues and adjacent normal tissues, 44 LAD tissues (received docetaxel-based chemotherapy)
Lymph node metastasis, tumour stage, overall survival rate, docetaxel resistance
[16]
CRC
Downregulated
Tissues from 96 CRC patients
Overall survival rate
[39]
CRC
Upregulated
45 rectal cancer tissues, 22 colon cancer tissues, adjacent noncancerous tissues
Oxaliplatin resistance, tumour growth
[40]
CRC(bioinf)
Downregulated
Tissues from 217 CRC patients
/
[41]
CRC
Upregulated
Tissues from 70 CRC patients
Lymph node metastasis
[42]
CRC
Upregulated
/
/
[14]
GC(bioinf)
Upregulated
Tissues from 90 GC patients, tissues from 90 pre-GC patients, tissues from 45 healthy controls
/
[45]
GC
Upregulated
Tissues from 119 GC patients
Overall survival rate
[46]
GC
Upregulated
93 GC tissues and adjacent normal tissues, 44 LAD tissues (received docetaxel-based chemotherapy)
Tumor growth
[47]
GC
Upregulated
Primary GC tissues
Lymphatic and distant metastasis
[15]
Glioma
Upregulated
168 glioma tissues and 21 normal contral tissues
WHO grade, KPS score, overall survival rate
[49]
Glioma
Upregulated
Tissues from 39 glioma patients
Tumour formation and growth
[50]
Glioma
Downregulated
Tissues from 24 glioma patients
/
[51]
Melonoma
Downregulated
Tissues from 309 melonoma patients
TNM stage, overall survial, progression-free survival
[19]
Melonoma
Downregulated
/
/
[56]
CLL
Downregulated
Peripheral blood from 80 CLL patients and healthy controls
/
[24]
CLL
Upregulated
Peripheral blood from 102 CLL patients and 40 healthy controls
Binet stage, lactate dehydrogenase (LDH) level, time for first treatment
[59]
ALL
Downregulated
/
Prognosis
[58]
AML
Downregulated
Bone marrow and peripheral blood from 40 AML patients and 35 healthy controls
/
[57]
BC
Upregulated
Tissues from BC patients
/
[60]
EC
Upregulated
60 EC tissues and adjacent normal tissues
Tumour size, TNM stage, lymph and distant metastasis
[11]
ATC
Upregulated
12 PTC tissues, 5 ATC tissues, 6 normal tissues
/
[61]
Osteosarcoma
Upregulated
/
/
[18]
OC
Upregulated
/
Tumour weight and volume
[62]
PC
Upregulated
216 PC tissues, 324 benign prostate tissues, 77 normal tissues
Tumour volume, tumour metastasis, mortality of severe
[63]
Table 2
The roles of miR-650 in various cancer cell lines
Cancer type
Cell lines
Upstream
Target
Roles
Function
References
HCC
HCC cell and LO2
/
LAST2
Tumour promoter
Promotes cell proliferation, migration and invasion; increases cell EMT
[30]
HCC
LO2, SK-HEP-1, HUH-7, LM6 and Li-7
AXIN1
/
Tumour promoter
Promotes cell proliferation, migration and invasion; increases cell EMT
[31]
HCC
THLE-2 (CRL-2706, ATCC)
/
ING4
Tumour promoter
Promotes cell proliferation
[34]
FLD/HCC
SMMC-7721 and BEL-7404
Benzo[a]pyrene
SOCS3/JAK/STAT3
Tumour promoter
Promotes cell motility
[32]
NSCLC
H23, H522, A549, H1299, SPC-A1, 16HBE, HEK293T
/
LATS2
Tumour promoter
Promotes cell proliferation, migration and invasion
[64]
NSCLC
A549, NCI-H460, MRC-5
/
ING4
Tumour promoter
Promotes cell proliferation and invasion
[13]
NSCLC
cell line H1299, 293T
LncRNA MEG3
SLC34A2
Tumour promoter
Promotes cell migration and invasion; strengthens stem cell-like characteristics
[37]
LAD
SPC-A1, NCI-H1299
/
ING4/Bcl-2/Bax
Tumour promoter
Promotes cell growth; enhances the resistance of LAD cells to docetaxel; reduces the sensitivity of LAD cells to docetaxel
[16]
CRC
DLD-1, HCT-8, HEK293T
/
AKT2/GSK3β/E-cadherin pathway
Tumour suppressor
Suppresses cell proliferation, migration and invasion
[39]
CRC
Caco2 and HT29
LncRNA MIR155HG
ANXA2
Tumour promoter
Promotes M2 macrophage polarization and cell proliferation, migration, invasion and oxaliplatin resistance
[40]
CRC(bioinf)
/
NF-κB
NF-κB signalling pathway
Tumour suppressor
/
[41]
CRC
SW480, HT29, SW620, LOVO
/
NDRG2
Tumour promoter
Promotes cell growth; suppresses cell differentiation and apoptosis
[42]
CRC
SW480, SW620, RKO, 320DM, 320HSR, NCI-H716, H508, CCD841CoN
/
RhoA/Rac1 GTPase + ING4/ERK/p38 MAPK
Tumour promoter
Promotes cell proliferation and migration; increases cell EMT
[14]
GC(bioinf)
/
/
/
Tumour promoter
/
[45]
GC
HGC-27, MKN-45 cells, HEK293T
PBX1
LATS2
Tumour promoter
Promotes cell proliferation, invasion and migration; suppresses cell apoptosis
[46]
GC
AGS, HGC-27, MGC-803, SGC-7901, GES-1
LncRNA DICER-AS1
CSR1
Tumour promoter
Promotes cell proliferation, migration; suppresses cell apoptosis
[47]
GC
SNU-16, NCI-N87
/
ING4
Tumour promoter
Promotes cell proliferation and tumorigenesis
[15]
Glioma
/
/
/
Tumour promoter
/
[49]
Glioma
U251, LN229, U373, A172, U87, NHA
NF-κB
RERG-PHLPP2/AKT + ERK + NF-κB
Tumour promoter
Promotes cell proliferation, autophagy, migration and invasion; increases EMT
[50]
Glioma
LN229, U87, U251, LN308, SNB19, H4
/
FAM83F
Tumour suppessor
Promotes cell proliferation
[51]
Melonoma
A375, MV3
LncRNA POU3F3
/
Tumour suppessor
Reduces MGMT-induced DTIC resistance; suppress cell proliferation
[19]
Melonoma
M21, SK-MEL-1, A2058, A375, HEMa-LP
LncRNA ZFPM2-AS1
Notch1
Tumour suppessor
Promotes cell proliferation and migration; suppresses cell apoptosis
[56]
CLL
NALM-6
host gene for IgLλ
CDK1 ING4 EBF3
Tumour suppessor
Suppresses proliferative capacity of B cells
[24]
CLL
CLL cells
/
NDRG2
Tumour promoter
Suppresses cell apoptosis
[59]
ALL
REH, NALM-6, UT-7
/
GATA2/EPOR
Tumour suppressor
/
[58]
AML
K562 cells
/
Gfi1
Tumour suppressor
Suppresses cell proliferation
[57]
BC
UIVC-IDC-6, -9, -10
22q11.2 gene
ING4 NDRG2
Tumour promoter
Increases cell EMT
[60]
EC
HEC-1B, HEC-1 A, Ishikawa, RL-952, hEEC
LncRNA MCTP1-AS1
SMAD7/TGF-β/SMAD pathway
Tumour promoter
Promotes cell proliferation, migration and invasion; increases EMT
[11]
ATC
TPC-1, CAL62, SW1736, 8505 C
/
PPP2CA
Tumour promoter
Promotes cell migration and invasion
[61]
Osteosarcoma
MG-63
/
NFκB + ING4/IL-6
Tumour promoter
/
[18]
OC
hTERT-OME, SCC-15, SCC-4, SCC-9, SCC-25, CAL-27, FaDu, 019
/
Gfi1
Tumour promoter
Promotes cell proliferation, migration and invasion
[62]
PC
PC3, DU145
/
CSR1
Tumour promoter
Promotes cell growth and DNA replication
[63]

Functions of miR-650 and the related mechanisms

In addition to the abberant expression files of miR-650, researchers have also observed variations in the biological functions of miR-650 through in vitro and in vivo experiments. Generally, miR-650 functions as an onco-miR or tumour suppressor by affecting biological processes such as cell proliferation and apoptosis. In the next section, we summarize the dual roles of miR-650 in tumours and the signalling pathways involved. Additionally, the comprehensive functions and related mechanisms of miR-650 in various tumours are presented in Figs. 1 and 2.

miR-650 functions as a tumour promoter

In parallel with the overexpression and clinical features discussed above, miR-650 also conferred more oncogenicity to tumour cells. In HCC, existing evidence indicates that upregulation of miR-650 modulates cell proliferation, apoptosis, migration and invasion [3032, 34] of cancer cells. In terms of the mechanism, miR-650 functions as an upstream signal of the large tumor suppressor kinase 2 gene (LATS2)/YAP (Ser127) signalling pathway and regulates its downstream target genes [30]. Moreover, Ye et al. [64] also revealed that miR-650 could serve as an onco-miR to potentiate cell growth and metastasis by directly targeting LATS2 in NSCLC formation and progression. Interestingly, Liu et al. [46] reported that miR-650 downregulation inhibited proliferation, expedited apoptosis and reduced the migration of HP + GC cells. Moreover, miR-650 mediated the Hippo pathway via the PBX1/miR-650/LATS axis. As a molecular sponge of Axin1, miR-650 enhanced the Wnt signalling pathway to facilitate tumour progression [31]. Furthermore, Ge et al. [32] found that miR-650 in benzo[a]pyrene-exposed cancer cells contributed to HCC metastasis by directly targeting suppressor of cytokine signalling 3 (SOCS3), and this inhibition modulated the activation of the Janus kinase (JAK)/ signal transducer and activator of transcription 3(STAT3) cascade. The study from Zhao et al. [37] suggested that miR-650 silencing inhibited the vital capacity and invasion ability of large-cell carcinomas (LCCs) and lung cancer stem cells (LCSCs) (H1299 cell lines) through the lncRNA maternally expressed gene 3(MEG3)/miR-650/solute carrier family 34 member 2 (SLC34A2) axis. Moreover, ING4 is also a vital gene targeted by miR-650 to facilitate cell proliferation and invasion and induce a stem cell-like state in LC [13, 16]. With regard to CRC, Zhou et al. [40] argued that MIR155HG, as an endogenous lncRNA, competed with annexin 2 (ANXA2) by combining with miR-650, thereby promoting M2 macrophage polarization and oxaliplatin resistance in CRC cells. In another study, decreased luciferase activity of miR-650 was observed with the 3’ untranslated region of N-myc downstream regulated gene 2 (NDRG2) inserted downstream of the luciferase gene, indicating that NDRG2 was directly targeted by miR-650 [65]. Notably, other researchers have observed that inhibition of miR-650 facilitates more apoptosis in CLL cells [59]. Further studies have demonstrated that NDRG2 expression can be significantly downregulated by miR-650 simultaneously with p53 aberrations [59]. It is worth mentioning that other researchers have found that constructed adenoviruses carrying the NDRG2 gene heightened p53-mediated apoptosis of HCC cells [66]. Moreover, CSR1 has also been reported to be a shared gene targeted by miR-650 in both GC [47] and prostate cancer [63]. In addition to cell proliferation and apoptosis, overexpression of miR-650 induces EMT of cancer cells [11, 14, 30, 49, 67], which is a key factor in promoting the metastasis of tumours [68]. Functional analyses in Jin’s study demonstrated that upregulated miR-650 expression heightened the migration of glioma cells through EMT promotion. Additionally, they found that miR-650 could inhibit glioma cell adhesion and promote autophagy. Mechanistically, NF-κB1 upregulated miR-650 expression by directly interacting with its promoter, and then the AKT/ extracellular regulated protein kinases (ERK) and NF-κB pathways were enhanced by miR-650 via the RAS-like, estrogen-regulated growth inhibitor (RERG)-PH domain and leucine rich repeat protein phosphatase 2 (PHLPP2) complex [50].
As a valued member of the ING family, ING4 has been revealed to function as a formidable tumour suppressor due to its significant role in the modification of chromatin modification, cell growth, cell invasion and vascularization [69, 70]. However, it has been revealed to be frequently decreased in various human tumours, and the variation in ING4 markedly contributes to cancer development. Interestingly, accumulating studies have suggested that ING4 is a downstream target of miR-650 in many types of cancer, such as HCC [34], LC [13, 16], CRC [14], GC [15], and BC [60]. Finally, You et al. [14] demonstrated that miR-650 could function as a tumour promoter and enhance the malignant phenotype of CRC. In terms of the mechanism, miR-650 targets ING4, leading to CRC progression promoted by the ERK/p38 mitogen-activated protein kinases (MAPK) pathway. Analogously, another study further suggested that miR-650 increased caspase-3-dependent cell apoptosis by regulating Bcl-2/Bax expression [16] via ING4. Interestingly, Tang et al. [13] revealed that miR-650 promotes NSCLC cell proliferation and migration through the ING4/Wnt-1/β-catenin pathway. Combined, these results suggest that ING4 is a significant regulator of the signalling pathways in the tumorigenic progress of miR-650 and provides promising biomarker and therapeutic target for human cancer.

miR-650 functions as a tumour suppressor

In contrast with the aforementioned investigations, miR-650 has also been reported to function as a tumour suppressor by arresting the cell cycle, inhibiting cell proliferation and weakening the malignant phenotype in tumours. In CRC, there is no consensus about the function of miR-650 thus far. Zhou et al. [39] found that miR-650 inhibited cell growth and migration by suppressing the AKT2/ Glycogen synthase kinase (GSK3β)/E-cadherin pathway. Another bioinformatic study revealed a similar conclusion [41, 42]. Therefore, to further explore the roles of miR-650, more related studies are needed. In addition, rescue experiments in Xu’s et al. [51] study showed that miR-650 could inhibit cell growth by targeting family with sequence similarity 83member F (FAM83F) in glioma. In addition, the lncRNA POU3F3/miR-650/MGMT pathway has been revealed to function critically in DTIC resistance in melanoma [19]. In terms of the mechanism, lncRNA POU3F3 works as a competitive RNA to combine with miR-650; therefore, MGMT expression rises to a higher extent [19] in melanoma cells. Additionally, Liu et al. [56] revealed that miR-650 contained cell proliferation and invasion while exerting on adverse effect on cell apoptosis in cutaneous melanoma via the lncRNA ZFPM2-AS1/miR-650/NOTCH1 axis in melanoma. An article published in Blood indicated that overexpression of miR-650 led to the decreased proliferative capacity of B cells [24]. Mechanistically, miR-650 targeted essential proteins in cell proliferation, namely, cyclin-dependent kinase (CDK1), ING4, and EBF3 in B cells. A cell transfection experiment with miR-650 revealed significant downregulation of signalling molecule levels of EBF3, ING4 and CDK1 by 67%, 64%, and 53%, respectively. This finding validates the correlation of these three proteins with miR-650 in B cells. Moreover, Mraz et al. [24] found that miR-650 was modulated by coupling expression with its homologous gene for immunoglobulin lambda. This observation is surprising because previous studies demonstrated that miR-650 has an independent expression of immunoglobulin [23].

Conclusions

In this review, we discussed the aberrant expression profiles, functions and underlying mechanisms of miR-650 in various cancer tissues and cell lines and focused on its upstream regulators and downstream target genes. As an important member of the miRNA family, miR-650 has been identified to play crucial roles in cancer genesis and progression via diversified signalling molecules, such as ING4, Gfi1, and LATS2, thereby regulating the proliferation, apoptosis, invasion and migration, EMT, and drug resistance of cancer cells. Taken together, accumulating evidence at present suggests that miR-650 can function as a tumour promoter in HCC, LC, GC, BC, EC, ATC, OC, PC, as a tumour suppressor in melanoma, and as both in CRC, glioma, and leukaemia. The discrepancy of its roles in cancer indicates that the functions of miR-650 are influenced by multiple factors, such as cancer cell types and the microecological environment. To fully understand its expressional functions, further studies of miR-650 molecular mechanisms are required.
This overview of the signalling pathways influenced by miR-650 provides us with a more comprehensive understanding of the complex association between miR-650 and human cancer. More significantly, valuable clues regarding the implications of miR-650 based on a constructed lncRNA-miRNA-mRNA molecular network will bring us more effective diagnostic and/or therapeutic strategies for various cancer patients. When arguing the suitability of this miRNA as a biomarker or therapeutic target, researchers need to discuss its availability and detectability in bodily fluid or exosomes and assess the stability in clinical tests.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
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 Bray F, Jemal A, Grey N, Ferlay J, Forman D. Global cancer transitions according to the Human Development Index (2008–2030): a population-based study. Lancet Oncol. 2012;13:790–801.PubMed Bray F, Jemal A, Grey N, Ferlay J, Forman D. Global cancer transitions according to the Human Development Index (2008–2030): a population-based study. Lancet Oncol. 2012;13:790–801.PubMed
2.
Zurück zum Zitat Farmer P, Frenk J, Knaul FM, Shulman LN, Alleyne G, Armstrong L, et al. Expansion of cancer care and control in countries of low and middle income: a call to action. Lancet. 2010;376:1186–93.PubMed Farmer P, Frenk J, Knaul FM, Shulman LN, Alleyne G, Armstrong L, et al. Expansion of cancer care and control in countries of low and middle income: a call to action. Lancet. 2010;376:1186–93.PubMed
3.
Zurück zum Zitat Huang L, Guo Z, Wang F, Fu L. KRAS mutation: from undruggable to druggable in cancer. Signal Transduct Target Ther. 2021;6:386.PubMedPubMedCentral Huang L, Guo Z, Wang F, Fu L. KRAS mutation: from undruggable to druggable in cancer. Signal Transduct Target Ther. 2021;6:386.PubMedPubMedCentral
6.
Zurück zum Zitat Kozomara A, Birgaoanu M, Griffiths-Jones S. miRBase: from microRNA sequences to function. Nucleic Acids Res. 2019;47:D155-62. Kozomara A, Birgaoanu M, Griffiths-Jones S. miRBase: from microRNA sequences to function. Nucleic Acids Res. 2019;47:D155-62.
7.
Zurück zum Zitat Bartel DP, Chen CZ. Micromanagers of gene expression: the potentially widespread influence of metazoan microRNAs. Nat Rev Genet. 2004;5:396–400.PubMed Bartel DP, Chen CZ. Micromanagers of gene expression: the potentially widespread influence of metazoan microRNAs. Nat Rev Genet. 2004;5:396–400.PubMed
8.
Zurück zum Zitat Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16:203–22.PubMed Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 2017;16:203–22.PubMed
9.
Zurück zum Zitat Li L, Yuan L, Luo J, Gao J, Guo J, Xie X. MiR-34a inhibits proliferation and migration of breast cancer through down-regulation of Bcl-2 and SIRT1. Clin Exp Med. 2013;13:109–17.PubMed Li L, Yuan L, Luo J, Gao J, Guo J, Xie X. MiR-34a inhibits proliferation and migration of breast cancer through down-regulation of Bcl-2 and SIRT1. Clin Exp Med. 2013;13:109–17.PubMed
10.
Zurück zum Zitat Qin X, Guo H, Wang X, Zhu X, Yan M, Wang X, et al. Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol. 2019;20:12.PubMedPubMedCentral Qin X, Guo H, Wang X, Zhu X, Yan M, Wang X, et al. Exosomal miR-196a derived from cancer-associated fibroblasts confers cisplatin resistance in head and neck cancer through targeting CDKN1B and ING5. Genome Biol. 2019;20:12.PubMedPubMedCentral
11.
Zurück zum Zitat Gao Q, Huang Q, Li F, Luo F. LncRNA MCTP1-AS1 regulates EMT process in endometrial cancer by targeting the miR-650/SMAD7 Axis. Onco Targets Ther. 2021;14:751–61.PubMedPubMedCentral Gao Q, Huang Q, Li F, Luo F. LncRNA MCTP1-AS1 regulates EMT process in endometrial cancer by targeting the miR-650/SMAD7 Axis. Onco Targets Ther. 2021;14:751–61.PubMedPubMedCentral
12.
Zurück zum Zitat Amano K. Survival and functional results after valve replacement. Kokyu To Junkan. 1990;38:951–61.PubMed Amano K. Survival and functional results after valve replacement. Kokyu To Junkan. 1990;38:951–61.PubMed
13.
Zurück zum Zitat Tang X, Ding Y, Wang X, Wang X, Zhao L, Bi H. miR-650 promotes non-small cell lung cancer cell proliferation and invasion by targeting ING4 through Wnt-1/β-catenin pathway. Oncol Lett. 2019;18:4621–8.PubMedPubMedCentral Tang X, Ding Y, Wang X, Wang X, Zhao L, Bi H. miR-650 promotes non-small cell lung cancer cell proliferation and invasion by targeting ING4 through Wnt-1/β-catenin pathway. Oncol Lett. 2019;18:4621–8.PubMedPubMedCentral
14.
Zurück zum Zitat You Q, Li H, Liu Y, Xu Y, Miao S, Yao G, et al. MicroRNA-650 targets inhibitor of growth 4 to promote colorectal cancer progression via mitogen activated protein kinase signaling. Oncol Lett. 2018;16:2326–34.PubMedPubMedCentral You Q, Li H, Liu Y, Xu Y, Miao S, Yao G, et al. MicroRNA-650 targets inhibitor of growth 4 to promote colorectal cancer progression via mitogen activated protein kinase signaling. Oncol Lett. 2018;16:2326–34.PubMedPubMedCentral
15.
Zurück zum Zitat Zhang X, Zhu W, Zhang J, Huo S, Zhou L, Gu Z, et al. MicroRNA-650 targets ING4 to promote gastric cancer tumorigenicity. Biochem Biophys Res Commun. 2010;395:275–80.PubMed Zhang X, Zhu W, Zhang J, Huo S, Zhou L, Gu Z, et al. MicroRNA-650 targets ING4 to promote gastric cancer tumorigenicity. Biochem Biophys Res Commun. 2010;395:275–80.PubMed
16.
Zurück zum Zitat Huang JY, Cui SY, Chen YT, Song HZ, Huang GC, Feng B, et al. MicroRNA-650 was a prognostic factor in human lung adenocarcinoma and confers the docetaxel chemoresistance of lung adenocarcinoma cells via regulating Bcl-2/Bax expression. PLoS ONE. 2013;8:e72615.PubMedPubMedCentral Huang JY, Cui SY, Chen YT, Song HZ, Huang GC, Feng B, et al. MicroRNA-650 was a prognostic factor in human lung adenocarcinoma and confers the docetaxel chemoresistance of lung adenocarcinoma cells via regulating Bcl-2/Bax expression. PLoS ONE. 2013;8:e72615.PubMedPubMedCentral
17.
Zurück zum Zitat Hernández-Aguilar JA, Cortina-Villar HS, García-Barrios LE, Castillo-Santiago M. Factors limiting formation of community forestry enterprises in the Southern Mixteca region of Oaxaca, Mexico. Environ Manage. 2017;59:490–504.PubMed Hernández-Aguilar JA, Cortina-Villar HS, García-Barrios LE, Castillo-Santiago M. Factors limiting formation of community forestry enterprises in the Southern Mixteca region of Oaxaca, Mexico. Environ Manage. 2017;59:490–504.PubMed
18.
Zurück zum Zitat Yun JH, Moon S, Lee HS, Hwang MY, Kim YJ, Yu HY, et al. MicroRNA-650 in a copy number-variable region regulates the production of interleukin 6 in human osteosarcoma cells. Oncol Lett. 2015;10:2603–9.PubMedPubMedCentral Yun JH, Moon S, Lee HS, Hwang MY, Kim YJ, Yu HY, et al. MicroRNA-650 in a copy number-variable region regulates the production of interleukin 6 in human osteosarcoma cells. Oncol Lett. 2015;10:2603–9.PubMedPubMedCentral
19.
Zurück zum Zitat Wu K, Wang Q, Liu YL, Xiang Z, Wang QQ, Yin L, et al. LncRNA POU3F3 contributes to dacarbazine resistance of human melanoma through the MiR-650/MGMT axis. Front Oncol. 2021;11:643613.PubMedPubMedCentral Wu K, Wang Q, Liu YL, Xiang Z, Wang QQ, Yin L, et al. LncRNA POU3F3 contributes to dacarbazine resistance of human melanoma through the MiR-650/MGMT axis. Front Oncol. 2021;11:643613.PubMedPubMedCentral
20.
Zurück zum Zitat Qu W, Jiang L, Hou G. Circ-AFF2/miR-650/CNP axis promotes proliferation, inflammatory response, migration, and invasion of rheumatoid arthritis synovial fibroblasts. J Orthop Surg Res. 2021;16:165.PubMedPubMedCentral Qu W, Jiang L, Hou G. Circ-AFF2/miR-650/CNP axis promotes proliferation, inflammatory response, migration, and invasion of rheumatoid arthritis synovial fibroblasts. J Orthop Surg Res. 2021;16:165.PubMedPubMedCentral
21.
Zurück zum Zitat Xu X, Chen H, Zhang Q, Xu J, Shi Q, Wang M. MiR-650 inhibits proliferation, migration and invasion of rheumatoid arthritis synovial fibroblasts by targeting AKT2. Biomed Pharmacother. 2017;88:535–41.PubMed Xu X, Chen H, Zhang Q, Xu J, Shi Q, Wang M. MiR-650 inhibits proliferation, migration and invasion of rheumatoid arthritis synovial fibroblasts by targeting AKT2. Biomed Pharmacother. 2017;88:535–41.PubMed
22.
Zurück zum Zitat Cummins JM, He Y, Leary RJ, Pagliarini R, Diaz LA Jr, Sjoblom T, et al. The colorectal microRNAome. Proc Natl Acad Sci USA. 2006;103:3687–92.PubMedPubMedCentral Cummins JM, He Y, Leary RJ, Pagliarini R, Diaz LA Jr, Sjoblom T, et al. The colorectal microRNAome. Proc Natl Acad Sci USA. 2006;103:3687–92.PubMedPubMedCentral
23.
Zurück zum Zitat Das S. Evolutionary origin and genomic organization of micro-RNA genes in immunoglobulin lambda variable region gene family. Mol Biol Evol. 2009;26:1179–89.PubMedPubMedCentral Das S. Evolutionary origin and genomic organization of micro-RNA genes in immunoglobulin lambda variable region gene family. Mol Biol Evol. 2009;26:1179–89.PubMedPubMedCentral
24.
Zurück zum Zitat Mraz M, Dolezalova D, Plevova K, Stano Kozubik K, Mayerova V, Cerna K, et al. MicroRNA-650 expression is influenced by immunoglobulin gene rearrangement and affects the biology of chronic lymphocytic leukemia. Blood. 2012;119:2110–3.PubMed Mraz M, Dolezalova D, Plevova K, Stano Kozubik K, Mayerova V, Cerna K, et al. MicroRNA-650 expression is influenced by immunoglobulin gene rearrangement and affects the biology of chronic lymphocytic leukemia. Blood. 2012;119:2110–3.PubMed
25.
Zurück zum Zitat Chen Z, Xie H, Hu M, Huang T, Hu Y, Sang N, et al. Recent progress in treatment of hepatocellular carcinoma. Am J Cancer Res. 2020;10:2993–3036.PubMedPubMedCentral Chen Z, Xie H, Hu M, Huang T, Hu Y, Sang N, et al. Recent progress in treatment of hepatocellular carcinoma. Am J Cancer Res. 2020;10:2993–3036.PubMedPubMedCentral
26.
Zurück zum Zitat Li C, Chen K, Liu X, Liu HT, Liang XM, Liang GL, et al. Analysis of clinicopathological characteristics and prognosis of young patients with hepatocellular carcinoma after hepatectomy. J Clin Transl Hepatol. 2020;8:285–91.PubMedPubMedCentral Li C, Chen K, Liu X, Liu HT, Liang XM, Liang GL, et al. Analysis of clinicopathological characteristics and prognosis of young patients with hepatocellular carcinoma after hepatectomy. J Clin Transl Hepatol. 2020;8:285–91.PubMedPubMedCentral
29.
Zurück zum Zitat Wang Y, Kui L, Wang G. Combination therapy for HCC: from CRISPR screening to the design of clinical therapies. Signal Transduct Target Ther. 2021;6:359.PubMedPubMedCentral Wang Y, Kui L, Wang G. Combination therapy for HCC: from CRISPR screening to the design of clinical therapies. Signal Transduct Target Ther. 2021;6:359.PubMedPubMedCentral
30.
Zurück zum Zitat Han LL, Yin XR, Zhang SQ. miR-650 promotes the metastasis and epithelial-mesenchymal transition of hepatocellular carcinoma by directly inhibiting LATS2 expression. Cell Physiol Biochem. 2018;51:1179–92.PubMed Han LL, Yin XR, Zhang SQ. miR-650 promotes the metastasis and epithelial-mesenchymal transition of hepatocellular carcinoma by directly inhibiting LATS2 expression. Cell Physiol Biochem. 2018;51:1179–92.PubMed
31.
Zurück zum Zitat Qin A, Wu J, Zhai M, Lu Y, Huang B, Lu X, et al. Axin1 inhibits proliferation, invasion, migration and EMT of hepatocellular carcinoma by targeting miR-650. Am J Transl Res. 2020;12:1114–22.PubMedPubMedCentral Qin A, Wu J, Zhai M, Lu Y, Huang B, Lu X, et al. Axin1 inhibits proliferation, invasion, migration and EMT of hepatocellular carcinoma by targeting miR-650. Am J Transl Res. 2020;12:1114–22.PubMedPubMedCentral
33.
Zurück zum Zitat Matboli M, Gadallah SH, Rashed WM, Hasanin AH, Essawy N, Ghanem HM, et al. mRNA-miRNA-lncRNA regulatory network in nonalcoholic fatty liver disease. Int J Mol Sci. 2021;22:6770.PubMedPubMedCentral Matboli M, Gadallah SH, Rashed WM, Hasanin AH, Essawy N, Ghanem HM, et al. mRNA-miRNA-lncRNA regulatory network in nonalcoholic fatty liver disease. Int J Mol Sci. 2021;22:6770.PubMedPubMedCentral
34.
Zurück zum Zitat Zeng ZL, Li FJ, Gao F, Sun DS, Yao L. Upregulation of miR-650 is correlated with down-regulation of ING4 and progression of hepatocellular carcinoma. J Surg Oncol. 2013;107:105–10.PubMed Zeng ZL, Li FJ, Gao F, Sun DS, Yao L. Upregulation of miR-650 is correlated with down-regulation of ING4 and progression of hepatocellular carcinoma. J Surg Oncol. 2013;107:105–10.PubMed
35.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.PubMed Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–49.PubMed
36.
Zurück zum Zitat Black RC, Khurshid H. NSCLC: an update of driver mutations, their role in pathogenesis and clinical significance. R I Med J (2013). 2015;98:25–8. Black RC, Khurshid H. NSCLC: an update of driver mutations, their role in pathogenesis and clinical significance. R I Med J (2013). 2015;98:25–8.
37.
Zurück zum Zitat Zhao Y, Zhu Z, Shi S, Wang J, Li N. Long non-coding RNA MEG3 regulates migration and invasion of lung cancer stem cells via miR-650/SLC34A2 axis. Biomed Pharmacother. 2019;120:109457.PubMed Zhao Y, Zhu Z, Shi S, Wang J, Li N. Long non-coding RNA MEG3 regulates migration and invasion of lung cancer stem cells via miR-650/SLC34A2 axis. Biomed Pharmacother. 2019;120:109457.PubMed
38.
Zurück zum Zitat Heist RS, Engelman JA. SnapShot: non-small cell lung cancer. Cancer Cell. 2012;21:448.e2.PubMed Heist RS, Engelman JA. SnapShot: non-small cell lung cancer. Cancer Cell. 2012;21:448.e2.PubMed
39.
Zurück zum Zitat Zhou C, Cui F, Li J, Wang D, Wei Y, Wu Y, et al. MiR-650 represses high-risk non-metastatic colorectal cancer progression via inhibition of AKT2/GSK3β/E-cadherin pathway. Oncotarget. 2017;8:49534.PubMedPubMedCentral Zhou C, Cui F, Li J, Wang D, Wei Y, Wu Y, et al. MiR-650 represses high-risk non-metastatic colorectal cancer progression via inhibition of AKT2/GSK3β/E-cadherin pathway. Oncotarget. 2017;8:49534.PubMedPubMedCentral
41.
Zurück zum Zitat Slattery ML, Mullany LE, Sakoda L, Samowitz WS, Wolff RK, Stevens JR, et al. The NF-κB signalling pathway in colorectal cancer: associations between dysregulated gene and miRNA expression. J Cancer Res Clin Oncol. 2018;144:269–83.PubMed Slattery ML, Mullany LE, Sakoda L, Samowitz WS, Wolff RK, Stevens JR, et al. The NF-κB signalling pathway in colorectal cancer: associations between dysregulated gene and miRNA expression. J Cancer Res Clin Oncol. 2018;144:269–83.PubMed
42.
Zurück zum Zitat Feng L, Xie Y, Zhang H, Wu Y. Down-regulation of NDRG2 gene expression in human colorectal cancer involves promoter methylation and microRNA-650. Biochem Biophys Res Commun. 2011;406:534–8.PubMed Feng L, Xie Y, Zhang H, Wu Y. Down-regulation of NDRG2 gene expression in human colorectal cancer involves promoter methylation and microRNA-650. Biochem Biophys Res Commun. 2011;406:534–8.PubMed
43.
Zurück zum Zitat Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.PubMed Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.PubMed
44.
Zurück zum Zitat Thrift AP, El-Serag HB. Burden of gastric cancer. Clin Gastroenterol Hepatol. 2020;18:534–42.PubMed Thrift AP, El-Serag HB. Burden of gastric cancer. Clin Gastroenterol Hepatol. 2020;18:534–42.PubMed
45.
Zurück zum Zitat Chen J, Wu L, Sun Y, Luo C, Chen X, Wu L, et al. Diagnostic value and clinical significance of circulating miR-650 and CA211 in detecting of gastric carcinoma. Oncol Lett. 2020;20:254.PubMedPubMedCentral Chen J, Wu L, Sun Y, Luo C, Chen X, Wu L, et al. Diagnostic value and clinical significance of circulating miR-650 and CA211 in detecting of gastric carcinoma. Oncol Lett. 2020;20:254.PubMedPubMedCentral
46.
Zurück zum Zitat Liu J, Wang L, Li J, Xu Y. Upregulation of microRNA-650 by PBX1 is correlated with the development of Helicobacter pylori-associated gastric carcinoma. Neoplasma. 2021;68:262–72.PubMed Liu J, Wang L, Li J, Xu Y. Upregulation of microRNA-650 by PBX1 is correlated with the development of Helicobacter pylori-associated gastric carcinoma. Neoplasma. 2021;68:262–72.PubMed
47.
Zurück zum Zitat An J, Guo X, Yan B. DICER-AS1 functions as competing endogenous RNA that targets CSR1 by sponging microRNA-650 and suppresses gastric cancer progression. J Int Med Res. 2021;49:3000605211041466.PubMed An J, Guo X, Yan B. DICER-AS1 functions as competing endogenous RNA that targets CSR1 by sponging microRNA-650 and suppresses gastric cancer progression. J Int Med Res. 2021;49:3000605211041466.PubMed
48.
Zurück zum Zitat Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7–33.PubMed Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71:7–33.PubMed
49.
Zurück zum Zitat Sun B, Pu B, Chu D, Chu X, Li W, Wei D. MicroRNA-650 expression in glioma is associated with prognosis of patients. J Neurooncol. 2013;115:375–80.PubMed Sun B, Pu B, Chu D, Chu X, Li W, Wei D. MicroRNA-650 expression in glioma is associated with prognosis of patients. J Neurooncol. 2013;115:375–80.PubMed
50.
Zurück zum Zitat Jin S, Li X, Dai Y, Li C, Wang D. NF-κB-mediated miR-650 plays oncogenic roles and activates AKT/ERK/NF-κB pathways by targeting RERG in glioma cells. Cell Oncol (Dordr). 2020;43:1035–48. Jin S, Li X, Dai Y, Li C, Wang D. NF-κB-mediated miR-650 plays oncogenic roles and activates AKT/ERK/NF-κB pathways by targeting RERG in glioma cells. Cell Oncol (Dordr). 2020;43:1035–48.
51.
Zurück zum Zitat Xu L, Yu QW, Fang SQ, Zheng YK, Qi JC. MiR-650 inhibits the progression of glioma by targeting FAM83F. Eur Rev Med Pharmacol Sci. 2018;22:8391–8.PubMed Xu L, Yu QW, Fang SQ, Zheng YK, Qi JC. MiR-650 inhibits the progression of glioma by targeting FAM83F. Eur Rev Med Pharmacol Sci. 2018;22:8391–8.PubMed
52.
Zurück zum Zitat Leonardi GC, Falzone L, Salemi R, Zanghì A, Spandidos DA, McCubrey JA, et al. Cutaneous melanoma: from pathogenesis to therapy (Review). Int J Oncol. 2018;52:1071–80.PubMedPubMedCentral Leonardi GC, Falzone L, Salemi R, Zanghì A, Spandidos DA, McCubrey JA, et al. Cutaneous melanoma: from pathogenesis to therapy (Review). Int J Oncol. 2018;52:1071–80.PubMedPubMedCentral
53.
Zurück zum Zitat Hartman RI, Lin JY. Cutaneous melanoma-A review in detection, staging, and management. Hematol Oncol Clin North Am. 2019;33:25–38.PubMed Hartman RI, Lin JY. Cutaneous melanoma-A review in detection, staging, and management. Hematol Oncol Clin North Am. 2019;33:25–38.PubMed
54.
Zurück zum Zitat Berk-Krauss J, Stein JA, Weber J, Polsky D, Geller AC. New systematic therapies and trends in cutaneous melanoma deaths among US whites, 1986–2016. Am J Public Health. 2020;110:731–3.PubMedPubMedCentral Berk-Krauss J, Stein JA, Weber J, Polsky D, Geller AC. New systematic therapies and trends in cutaneous melanoma deaths among US whites, 1986–2016. Am J Public Health. 2020;110:731–3.PubMedPubMedCentral
55.
Zurück zum Zitat Mason R, Au L, Ingles Garces A, Larkin J. Current and emerging systemic therapies for cutaneous metastatic melanoma. Expert Opin Pharmacother. 2019;20:1135–52.PubMed Mason R, Au L, Ingles Garces A, Larkin J. Current and emerging systemic therapies for cutaneous metastatic melanoma. Expert Opin Pharmacother. 2019;20:1135–52.PubMed
56.
Zurück zum Zitat Liu W, Hu X, Mu X, Tian Q, Gao T, Ge R, et al. ZFPM2-AS1 facilitates cell proliferation and migration in cutaneous malignant melanoma through modulating miR-650/NOTCH1 signaling. Dermatol Ther. 2021;34:e14751.PubMed Liu W, Hu X, Mu X, Tian Q, Gao T, Ge R, et al. ZFPM2-AS1 facilitates cell proliferation and migration in cutaneous malignant melanoma through modulating miR-650/NOTCH1 signaling. Dermatol Ther. 2021;34:e14751.PubMed
57.
Zurück zum Zitat Yuan C, Xu L, Du P, Pang J. miRNA-650 exerts anti-leukemia activity by inhibiting cell proliferation through Gfi1 targeting. Tumori. 2018;104:369–74.PubMed Yuan C, Xu L, Du P, Pang J. miRNA-650 exerts anti-leukemia activity by inhibiting cell proliferation through Gfi1 targeting. Tumori. 2018;104:369–74.PubMed
58.
Zurück zum Zitat Gaine ME, Sharpe DJ, Smith JS, Colyer HAA, Hodges VM, Lappin TR, et al. GATA2 regulates the erythropoietin receptor in t(12;21) ALL. Oncotarget. 2017;8:66061–74.PubMedPubMedCentral Gaine ME, Sharpe DJ, Smith JS, Colyer HAA, Hodges VM, Lappin TR, et al. GATA2 regulates the erythropoietin receptor in t(12;21) ALL. Oncotarget. 2017;8:66061–74.PubMedPubMedCentral
59.
Zurück zum Zitat Yang YQ, Tian T, Zhu HY, Liang JH, Wu W, Wu JZ, et al. NDRG2 mRNA levels and miR-28-5p and miR-650 activity in chronic lymphocytic leukemia. BMC Cancer. 2018;18:1009.PubMedPubMedCentral Yang YQ, Tian T, Zhu HY, Liang JH, Wu W, Wu JZ, et al. NDRG2 mRNA levels and miR-28-5p and miR-650 activity in chronic lymphocytic leukemia. BMC Cancer. 2018;18:1009.PubMedPubMedCentral
60.
Zurück zum Zitat Lango-Chavarría M, Chimal-Ramírez GK, Ruiz-Tachiquín ME, Espinoza-Sánchez NA, Suárez-Arriaga MC, Fuentes-Pananá EM. A 22q11.2 amplification in the region encoding microRNA-650 correlates with the epithelial to mesenchymal transition in breast cancer primary cultures of Mexican patients. Int J Oncol. 2017;50:432–40.PubMedPubMedCentral Lango-Chavarría M, Chimal-Ramírez GK, Ruiz-Tachiquín ME, Espinoza-Sánchez NA, Suárez-Arriaga MC, Fuentes-Pananá EM. A 22q11.2 amplification in the region encoding microRNA-650 correlates with the epithelial to mesenchymal transition in breast cancer primary cultures of Mexican patients. Int J Oncol. 2017;50:432–40.PubMedPubMedCentral
61.
Zurück zum Zitat Orlandella FM, Mariniello RM, Iervolino PLC, Imperlini E, Mandola A, Verde A, et al. miR-650 promotes motility of anaplastic thyroid cancer cells by targeting PPP2CA. Endocrine. 2019;65:582–94.PubMed Orlandella FM, Mariniello RM, Iervolino PLC, Imperlini E, Mandola A, Verde A, et al. miR-650 promotes motility of anaplastic thyroid cancer cells by targeting PPP2CA. Endocrine. 2019;65:582–94.PubMed
62.
Zurück zum Zitat Ningning S, Libo S, Chuanbin W, Haijiang S, Qing Z. MiR-650 regulates the proliferation, migration and invasion of human oral cancer by targeting growth factor independent 1 (Gfi1). Biochimie. 2019;156:69–78.PubMed Ningning S, Libo S, Chuanbin W, Haijiang S, Qing Z. MiR-650 regulates the proliferation, migration and invasion of human oral cancer by targeting growth factor independent 1 (Gfi1). Biochimie. 2019;156:69–78.PubMed
63.
Zurück zum Zitat Zuo ZH, Yu YP, Ding Y, Liu S, Martin A, Tseng G, et al. Oncogenic activity of miR-650 in prostate cancer is mediated by suppression of CSR1 expression. Am J Pathol. 2015;185:1991–9.PubMedPubMedCentral Zuo ZH, Yu YP, Ding Y, Liu S, Martin A, Tseng G, et al. Oncogenic activity of miR-650 in prostate cancer is mediated by suppression of CSR1 expression. Am J Pathol. 2015;185:1991–9.PubMedPubMedCentral
64.
Zurück zum Zitat Ye Y, Zhuang J, Wang G, He S, Ni J, Xia W, et al. microRNA-605 promotes cell proliferation, migration and invasion in non-small cell lung cancer by directly targeting LATS2. Exp Ther Med. 2017;14:867–73.PubMedPubMedCentral Ye Y, Zhuang J, Wang G, He S, Ni J, Xia W, et al. microRNA-605 promotes cell proliferation, migration and invasion in non-small cell lung cancer by directly targeting LATS2. Exp Ther Med. 2017;14:867–73.PubMedPubMedCentral
65.
Zurück zum Zitat O’Dwyer L, Lamberton F, Matura S, Tanner C, Scheibe M, Miller J, et al. Reduced hippocampal volume in healthy young ApoE4 carriers: an MRI study. PLoS ONE. 2012;7:e48895.PubMedPubMedCentral O’Dwyer L, Lamberton F, Matura S, Tanner C, Scheibe M, Miller J, et al. Reduced hippocampal volume in healthy young ApoE4 carriers: an MRI study. PLoS ONE. 2012;7:e48895.PubMedPubMedCentral
66.
Zurück zum Zitat Cao W, Zhang JL, Feng DY, Liu XW, Li Y, Wang LF, et al. The effect of adenovirus-conjugated NDRG2 on p53-mediated apoptosis of hepatocarcinoma cells through attenuation of nucleotide excision repair capacity. Biomaterials. 2014;35:993–1003.PubMed Cao W, Zhang JL, Feng DY, Liu XW, Li Y, Wang LF, et al. The effect of adenovirus-conjugated NDRG2 on p53-mediated apoptosis of hepatocarcinoma cells through attenuation of nucleotide excision repair capacity. Biomaterials. 2014;35:993–1003.PubMed
67.
Zurück zum Zitat Schwartz HI, Vingiano W, Perez CB. Autonomy and the right to refuse treatment: patients’ attitudes after involuntary medication. Hosp Commun Psychiatry. 1988;39:1049–54. Schwartz HI, Vingiano W, Perez CB. Autonomy and the right to refuse treatment: patients’ attitudes after involuntary medication. Hosp Commun Psychiatry. 1988;39:1049–54.
68.
Zurück zum Zitat Serrano-Gomez SJ, Maziveyi M, Alahari SK. Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications. Mol Cancer. 2016;15:18.PubMedPubMedCentral Serrano-Gomez SJ, Maziveyi M, Alahari SK. Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications. Mol Cancer. 2016;15:18.PubMedPubMedCentral
69.
Zurück zum Zitat Cui S, Gao Y, Zhang K, Chen J, Wang R, Chen L. The emerging role of inhibitor of growth 4 as a tumor suppressor in multiple human cancers. Cell Physiol Biochem. 2015;36:409–22.PubMed Cui S, Gao Y, Zhang K, Chen J, Wang R, Chen L. The emerging role of inhibitor of growth 4 as a tumor suppressor in multiple human cancers. Cell Physiol Biochem. 2015;36:409–22.PubMed
70.
Zurück zum Zitat Yan R, He L, Li Z, Han X, Liang J, Si W, et al. SCF(JFK) is a bona fide E3 ligase for ING4 and a potent promoter of the angiogenesis and metastasis of breast cancer. Genes Dev. 2015;29:672–85.PubMedPubMedCentral Yan R, He L, Li Z, Han X, Liang J, Si W, et al. SCF(JFK) is a bona fide E3 ligase for ING4 and a potent promoter of the angiogenesis and metastasis of breast cancer. Genes Dev. 2015;29:672–85.PubMedPubMedCentral
Metadaten
Titel
Functions and underlying mechanisms of miR-650 in human cancers
verfasst von
Yuanshuai Su
Qiuxian Zheng
Lingxiao Zhu
Xinyu Gu
Juan Lu
Lanjuan Li
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/s12935-022-02551-9

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.