Skip to main content
Technology in Cancer Research & Treatment logoLink to Technology in Cancer Research & Treatment
. 2022 Oct 26;21:15330338221134385. doi: 10.1177/15330338221134385

Depicting the Implication of miR-378a in Cancers

Yuelan Qin 1,*, Renba Liang 2,*, Pingan Lu 3, Lin Lai 1, Xiaodong Zhu 1,4,5,
PMCID: PMC9608056  PMID: 36285472

Abstract

MicroRNA-378a (miR-378a), including miR-378a-3p and miR-378a-5p, are encoded in PPARGC1B gene. miR-378a is essential for tumorigenesis and is an independent prognostic biomarker for various malignant tumors. Aberrant expression of miR-378a affects several physiological and pathological processes, including proliferation, apoptosis, tumorigenesis, cancer invasion, metastasis, and therapeutic resistance. Interestingly, miR-378a has a dual functional role in either promoting or inhibiting tumorigenesis, independent of the cancer type. In this review, we comprehensively summarized the role and regulatory mechanisms of miR-378a in cancer development, hoping to provide a direction for its potential use in cancer therapy.

Keywords: miR-378a, tumorigenesis, migration, invasion, proliferation, targets, clinical applications

Introduction

Micro RNAs (miRNAs) are small non-coding RNAs (19-24 nucleotides) produced from an endogenous transcript having a local hairpin structure with the help of RNase III-type enzyme.1 miRNAs can repress the translation of target proteins by cleaving or clinging to the 3′ untranslated region (UTR) of the corresponding messenger RNAs (mRNAs).2 The first miRNA was discovered over 20 years ago in mammals.3 Based on their genomic origin, miRNAs are divided into intergenic, intronic, and exonic.4 They are essential regulators of various critical biological processes, such as cell differentiation,5,6 apoptosis,7,8 proliferation,9 cell division,10 protein secretion,11 and viral infection.12 Recently, several studies have shown that the aberrant expression of miRNA is tightly associated with the progression of human diseases, especially cancers,13 as they can suppress or promote tumor growth by regulating the target gene mRNA.14-16

The miRNA miR-378a is one such essential tumor-regulating miRNA. It is located on chromosome 5q32 and embedded within the first intron of the PPARGC1β gene, which encodes the PGC1β protein. miR-378a is divided into miR-378a-3p (usually identified as miR-378) and miR-378a-5p (usually identified as miR-378*), which are the guide and passenger strands of miR-378a, respectively.17 Both miR-378a-3p and miR-378a-5p are synchronously transcribed with PGC1β,18-20 although there is an exception. Recently, it has been demonstrated that the over-expression (Table 1) or under-expression (Table 2) of miR-378a regulates the expression of its target genes and affects the proliferation,21 invasion, and metastasis22 of cancers. For example, low miR-378a expression is usually correlated to a significantly poor overall survival (OS)23 in colorectal cancer patients and associated with significantly decreased OS and disease-free survival (DFS) in gastric cancer patients.24 However, its expression and role in different cancers, such as lung25 and cervical cancers,26 is still controversial. miR-378a was identified as an onco-RNA in various tumors, including lung cancer,22,25,27-29 ovarian cancer,30-32 cervical cancer,33 nasopharyngeal carcinoma,34 melanoma,35 osteosarcoma,36 cholangiocarcinoma,37 acute myeloid leukemia,38 chronic myeloid leukemia,21 and Burkitt lymphoma.39 In contrast, it acted as an tumor suppressor in gastric cancer,40 colorectal cancer,41 liver cancer,42 glioblastoma,43 prostate cancer,44 breast cancer (BC),45 medulloblastoma (MB),46 pituitary adenoma (PA),47 oral squamous cell carcinomas,48 bladder cancer,49 esophageal carcinoma,50 rhabdomyosarcoma (RMS),51 and retinoblastoma (RB).52 Compared with normal tissues and no lymph node metastasis cancer tissues, miR-378a was upregulated in cervical cancer tissues, especially in CIN III and lymph node metastasis cervical cancer.33 Additionally, a study of 120 cholangiocarcinoma tissues and adjacent noncancerous tissues found that miR-378a expression increased with the development of the TNM (T-tumor, N-nodes, M-metastasis) stages.37 Positive miR-378a expression is also tightly linked to a low OS in cholangiocarcinoma patients.37 However, in colorectal cancer patients, miR-378a is downregulated and low miR-378a correlated with poor OS,4 suggesting an inhibiting effect of miR-378a.24 Thus, miR-378a can drive cancer cell proliferation,29,53 invasion, and migration27,32 in some tumors, while inhibiting these processes in other ftumors.23,24,32,54,55 Further, a study on the radiation response in a glioblastoma xenograft model found that the overexpression of miR-378a was associated with enhanced local tumor response to radiotherapy by increasing the vascular density and perfusion, thereby prolonging the survival of tumor-bearing hosts and acting as a novel therapeutic intervention.43 To summarize, the abnormal expression of miR-378a is crucial for the occurrence and development of malignant tumors. Therefore, it is important to understand the functions of miR-378a in tumor biology to enhance the efficiency of early screening and diagnosis of cancer, strengthen anticancer therapy, and develop novel targeted methods for tumor therapy. Hence, this review focuses on the roles and targets of miR-378a in different cancers to elucidate the specific molecular mechanisms of miR-378a during tumor progression.

Table 1.

miR-378a Plays Tumorigenic Roles as an Oncogene.

Tumor Sample Type Expression of MiR-378a in Tumor sample Role of MiR-378a Upregulation in Vitro Role of miR-378a Upregulation in Vivo Reference
Lung cancer Cancer tissue and cell Lung cancer tissue with brain metastasis (BM)-↓, BM + ↑; A549↑ Cell survival↑, migration and invasion↑, angiogenesis↑ Growth↑, angiogenesis↑ 22
Cell A549↑, SK-LU-1↓ Invasion↑ Metastasis↑, angiogenesis↑ 27
Cancer tissue and cell Lung cancer tissue↑; A549↑, H1299↑, H460↑, H520↑, H1975↑, PC9↑ Cell growth↑, apoptosis↓ 28
Cell SK-MES-1↓, A549↓ NCI-H460↑, NCI-H292↑ Proliferation↑, migration↑, angiogenesis↑ Growth ↑, metastasis↑ 29
Serum Serum exosomal miR-378a↑ 57
Cell Proliferation↑, migration↑ 59
Cancer tissue Lung cancer tissue↑ Proliferation↑ 60
Cell A549↑, A549/CDDP↓, Anip973↑, Anip973/CDDP↓ CDDP sensitivity↑, apoptosis↑ Growth↓ 25
OC Cell SKOV3↑, CAOV3↑ Proliferation↑, migration↑, invasion↑, angiogenesis↑ 32
Cell SKOV3↑, A2780↑, H281↓, H252↓, H254↓ 62
Cell A2780↑, CAOV3↑, OVCAR3↓ - - 63
CC Cancer tissue Normal tissue↓; cancer tissue↑ Migration↑, invasion↑ Metastasis↑ 33
Cancer tissue and cell CC tissue↑; HeLa↑, C33A↑, SiHa↑, CaSKi↑, End1/E6E7↓ Growth↑,apoptosis↓, cell cycle progression↑, Wnt/β-catenin pathway↑ 65
Serum and cancer tissue Serum of CC patients↓; CC tissue↓ Proliferation↓ Growth↓ 26
Cell C33A↓, ME-1800↓ Proliferation↓, Migration↓, invasion↓ 64
Melanoma Cancer tissue Melanoma tissue↑, lymph node metastasis↑ Migration↑, invasion↑, EMT↑ Growth↑ 35
Melanoma Cancer tissue Melanoma tissue↑ Proliferation↑, migration↑, angiogenesis↑ Angiogenesis↑ 67
NPC Cell 5-8F↑, 6-10B↑, CNE1↑ CNE2↑ Proliferation↑, migration and invasion↑ Growth↑ 34
Osteosarcoma Cancer tissue and cell Osteosarcoma tissue↑; Saos2↑, U2OS↑, MG-63↑, HOS↑ Proliferation↑ - 71
Cholangiocarcinoma Cancer tissue and cell Cholangiocarcinoma tissue↑; QBC939↑, RBE↑ Proliferation↑, migration and invasion↑ - 37
Renal carcinoma Serum RCC patients↑, patients after radical nephrectomy↓ - - 73
Serum RCC patients↑ 74,75
BL Cell ST486↑, Ramos↑, CA46↑DG75↑ Growth↑ 39
AML Cancer sample and cell AML samples↓; bone marrow mononuclear cells (BMNCs)↑ 38
CML BMMNCs BMMNCs↑ Proliferation↑, apoptosis ↓, drug resistance to 5-FU↑, ability of stem cell sphere formation↑ - 21

↑indicates upregulation (in case of expression level) or promoting (in case of functional consequences).

↓indicates downregulation (in case of expression level) or suppressing (in case of functional consequences).

Table 2.

miR-378a Plays Inhibiting Roles by Acting as a Tumor Suppressor.

Tumor Sample Type Expression of MiR-378a in Tumor Sample Role of miR-378a Upregulation in Vitro Role of miR-378a Upregulation in Vivo Reference
GC Cancer tissue and cell GC tissue↓; BGC-823↓, MKN-28↓, MGC-803↓, SGC-7901↓, AGS↓ Invasion↓, migration↓, EMT↓ 24
Cancer tissue and cell GC tissue↓; SGC-7901↓, AGS↓, MGC-803↓ Proliferation↓, G2/M phases↑ 40
Cancer tissue and cell GC tissue↓; KATO III↑, HGC-27↓, SNU-1↓ Proliferation↓, delay the cell cycle 54
Cancer tissue GC tissue↓ 82
Serum and cancer tissue Serum level of GC patients↑, GC tissue↓ - - 83
CRC Cancer tissue and cell CRC tissue↓; SW620↓, SW480↓, HT29↓, HCT116↓ Progression↓, invasion↓ Growth↓ 23
Cancer tissue and cell CRC tissue↓; HCT116↓, HT29↓, DLD-1↓, LOVO↓, SW620↓, SW480↓ Proliferation↓, cell of G0/G1 phase↑ and S-phase↓, cisplatin-induced apoptosis↑ Growth↓ 41
Cancer tissue and cell CRC tissue↓; LoVo↓, CaCo2↓, SW1116↓, SW480↓, HCT-116↓ Proliferation↓, apoptosis↑ 55
Cancer tissue and cell CRC and cell↓; HT29↓, HCT116↓, SW480↓, SW1116↓, SW620↓, LOVO↓ Proliferation↓, apoptosis↑ 86
Plasma Plasma level of CRC patient↓ 88
Cancer tissue CRC tissue↓ Invasion↓ 91
Cancer tissue and cell CRC tissue↓; HT-29↓, SW620↓, HT55↓ Growth↓ 92
Cancer tissue and cell CRC tissue↓; LoVo↓, CaCo2↓, SW1116↓, SW480↓, HCT-116↓ Proliferation↓, apoptosis↑, migration and invasion↓ 93
Proliferation↓, migration and invasion↓ 94
Liver cancer Cell Proliferation↓ Growth↓ 42
Cancer tissue and cell Liver cancer tissue↓; HepG2↓, HuH7↓, SMMC-7721↓, Li-7↓, PLC/PRF5↓, SK-Hep-1↓, MHCC97L↓, and MHCC97↓ Growth↓, migration and invasion↓, PLAGL2/β-catenin signaling↓ Growth↓, distant metastasis↓ 96
Cancer tissue and cell HCC tissue↓; SMMC-7721↓, MHCC-97H↓, MHCC-97L↓, HCCLM3↓ Angiogenesis↓, NF-κB signaling↓ Angiogenesis↓ 97
Cancer tissue Liver cancer tissue↓ Proliferation↓, invasion ↓, sorafenib-based chemotherapies↑ 98
Cancer tissue and cell Migration and invasion↓ 101
Cancer cell Sorafenib resistance cell↓ Response to sorafenib↑ 102
Cancer tissue HCC tissue↓ HCC cell viability↓, apoptosis↑, migration and invasion↓ - 103
Glioma Cancer tissue and cell Glioma tissue↓; SHG44↓, A172↓, LN229↓, LN18↓, T98↓ Migration and invasion↓, EMT↓ Growth↓ 108
Cancer tissue and cell Glioma tissue↓; U87↓, U251↓, MT330↓, SJ-G2↓ Migration and invasion↓ 109
Cancer tissue and cell Glioma tissue↓; U87MG↓, MT-330↓ Proliferation↓, apoptosis↑, migration and invasion↓ 110
PCa Cancer tissue and cell PCa tissue↓; LNCAP↓, PC-3M1E8↓, PC-3M↓, PC-3M2B4↓, du145↓ Proliferation↓, apoptosis↑, migration and invasion↓, cell in G1/S phase↑ Growth↓ 44
Cancer tissue PCa tissue↓ 113
Cancer tissue PCa tissue↓ 115
Cell 22Rv1↓, LNCaP↓, PC3↓, DU145↓ Proliferation↓, glucose metabolism↓ 114
BC Cancer tissue from mouse model and BC cell of human BC tissue↓; MDA-231↓ Migration and invasion↓ 45
Cell Tamoxifen-resistant MCF-7 cells↓, long-term estrogen-deprived MCF-7 cells↓ Growth↓ and apoptosis↑ 116
Proliferation↓ 118
MB Cancer tissue MB tissue↓ Proliferation↓, apoptosis↑ 46
PA Cancer tissue PA tissue↓ Proliferation↓, migration↓ 47
OSCC Cancer tissue OSCC tissue↓ 121
Migration and invasion↓, EMT↓ Metastasis↓ 48
Cancer tissue and cell OSCC tissue↓; Tca8113↓, CAL-27↓ Proliferation↓, apoptosis↑ 121
Bladder cancer Cancer tissue Bladder cancer tissue↓ 49
Cancer tissue Bladder cancer tissue↓ Cell viability↓, migration and invasion↓ 123
ESCC Cancer tissue and cell ESCC tissue↓; TE-1↓, KYSE-150↓, Eca-109↓, TE-8 Proliferation↓, migration and invasion↓ 50
Cancer tissue and cell ESCC tissue↓; EC109↓, KYSE150↓ Proliferation↓, apoptosis↑, cells in G1 phase↑, migration and invasion↓ 124
Cancer tissue and cell ESCC tissue↓; Eca109↓, KYSE150↓, KYSE30↓, KYSE450↓, KYSE70↓ Proliferation↓, invasion↓ 126
RMS Cancer tissue and cell RMS tissue↓; ARMS RH30↓, ERMS RD↓ Apoptosis↑, migration↓ 51
RB Cancer tissue RB tissue↓ Cell viability↓, apoptosis↑ Growth↓ 52

↑indicates upregulation (in case of expression level) or promoting (in case of functional consequences).

↓indicates downregulation (in case of expression level) or suppressing (in case of functional consequences).

Dual Function of miR-378a in Cancer Development

Growing research studies have elucidated the critical role of miR-378a in cancer progression, and most studies focused on the specific targets of miR-378a in tumor cell proliferation, migration, invasion, and regulatory pathways. Here, we summarize the roles and targets of miR-378a in tumorigenesis. Interestingly, both oncogenic (Table 1) and tumor-suppressive (Table 2) roles have been reported for miR-378a, implicating its dual function in cancer progression. By regulating downstream target genes, miR-378a promotes (Table 3) or inhibits tumors (Table 4), while it is regulated by its upstream regulators (Figure 1).

Table 3.

miR-378a Functions as an Onco-miRNA in Cancers by Regulating the Downstream Targets.

Cancer Type Expression of Downstream Target When miR-378a is Upregulated Effects of Changes in Target Genes Reference
Lung cancer SOX7↓ Promote proliferation, invasion, migration and EMT but inhibited cell apoptosis 59
RBX1↓ Promote invasion 27
HMOX1↓ Promote proliferation, migration and angiopoiesis 29
FOXG1↓ Promote proliferation and cell cycle distribution at G0/G1 phase but attenuated cell apoptosis 28
SCLU↓ Promote chemoresistance of cisplatin and apoptosis 25
CDK4↓ Inhibit proliferation 60
CDK6↓ Inhibit proliferation 60
TUSC2↓ Promote growth 61
OC ALCAM↑ A marker for better response to antiangiogenic therapy 62
EHD1↓ A marker for worse response to antiangiogenic therapy 62
SMAD4↓ Promote proliferation, migration, invasion, and angiogenesis 32
CC CPEB3↓ Promote proliferation, migration, invasion and EMT but decrease apoptosis 64
ATG12↓ Promote migration and invasion 33
ST7L↓ Promote proliferation by regulating the cell cycle process but inhibit apoptosis and Wnt/β-catenin pathway 65
Melanoma SUFU↓ Unknown 67
FUS-1↓ Unknown 67
KLF9↓ Unknown 67
STAMBP↓ Unknown 67
HOXD10↓ Unknown 67
NPC TOB2↓ Promote proliferation, migration, invasion in vitro and tumor growth vivo 71
Osteosarcoma KLF9↓ Promote proliferation 36
BYSL↓ Inhibit EMT and invasion 72
Renal cell carcinoma POLR2A↓ Promote apoptosis 77
RUNX2↓ Promote apoptosis 77
BL IRAK4↓ Promote cell growth 39
MNT↓ Promote cell growth 39
AML ZBTB20↑ Promote growth and invasion 79
FUS1↓ Promote proliferation 80
CML FUS1↓ Promoted proliferation 21

↑indicates upregulation; ↓indicates downregulation.

Table 4.

miR-378a Functions as a Suppressor in Various Cancers by Regulating the Downstream Targets.

Cancer Type Expression of Downstream Target When miR-378a is Upregulated Effects of Changes in Target Genes Reference
GC BMP2↓ Inhibit migration, invasion and EMT 24
MAPK1↓ Inhibit proliferation, migration, invasion and cell cycle progression 84
VEGF↓ Unknown 85
CRC MED19↓ Inhibit proliferation, migration, invasion and EMT, decrease cells in S phase and increase cells in G0 phase 91
IGF1R↓ Unknown 86
CDC40↓ Inhibit cell growth 41
Vimentin↓ Unknown 23
BRAF↓ Inhibit proliferation and induce apoptosis 55
SDAD1↓ Inhibit proliferation, migration and invasion 93
KISS1↑ Inhibit proliferation, migration and invasion 94
Liver cancer VEGFR↓ Unknown 98
PDGFRβ↓ Unknown 98
C-Raf↓ Unknown 98
FUS↓ Unknown 100
VEGF↓ Inhibit migration, invasion and proliferation 101
IGF1R↓ Promote sorafenib sensitivity 102
Glioma IRG1↓ Inhibit metastasis and EMT 108
TSPAN17↓ Inhibit proliferation migration, invasion and decreased apoptosis 110
SUFU↓ Increase cell survival, tumor growth and angiogenesis 112
FUS-1↓ Increase cell survival, tumor growth and angiogenesis 112
PCa GLUT1↓ Inhibit glycolytic metabolism and decrease cell survival 114
KLK2↑ Unknown 113
KLK4↑ Unknown 113
MAPK↓ Inhibit proliferation, invasion and migration, induce apoptosis and cause cell cycle arrest 44
BC YY1↓ Unknown 117
DCTPP1↓ Inhibit proliferation, induce apoptosis and activate DNA Repair mediated signaling pathway in vitro, repress tumorigenesis in vivo 118
RUNX1↓ Inhibit migration 45
SUFU↓ Decrease apoptosis, increase chemotherapy-resistant 119
KLK4↓ Unknown 120
MB UHRF1↓ Inhibit proliferation and apoptosis 46
PA RNF31↓ Inhibit proliferation and migration 47
OSCC KLK4↓ Inhibit migration and invasion 48
GLUT1↓ Inhibit glycolysis and cell proliferation 122
NCAPG↓ Inhibit proliferation and decrease apoptosis 123
ESCC Rab10↓ Inhibit proliferation, invasion and migration 125
GLUT1↓ Inhibit proliferation, invasion and induce apoptosis 126
SPHK1↓ Inhibit proliferation and invasion 127
RMS IGF1R↓ Unknown 51
RB FOXG1↓ Inhibit proliferation and promote apoptosis 52

↑indicates upregulation; ↓indicates downregulation

Figure 1.

Figure 1.

The regulatory factors upstream of miR-378a and the downstream target genes regulated by miR-378a in human cancer. miR-378a can be targeted and regulated by lncRNAs and circRNAs, thereby affecting its downstream gene expression and playing an important role in tumors.

Upregulation of miR-378a Promotes Malignant Transformation and Progression

Lung Cancer

Lung cancer is a common malignant tumor with a high mortality rate.56 Despite advances in cancer therapy, the OS rate of patients with advanced lung cancer is low. The expression of miR-378a both in tumor tissues and cell lines was significantly higher than in adjacent normal tissues or cells, and upregulation of miR-378a promoted the tumor progression.22,25,27-29 Further, miR-378a was significantly overexpressed in serum exosomes of non-small cell lung cancer (NSCLC) patients. The aberrant expression of miR-378a was closely associated with an advanced TNM stage, positive metastasis, negative therapeutic response, and poor OS.25,57 miR-378a can promote NSCLC cell migration, invasion, and tumor angiogenesis.22,27 Moreover, the circular RNA has-circ-0007059 was identified as an upstream regulatory factor of miR-378a, which can inhibit the proliferation and epithelial-mesenchymal transition (EMT) of A549 and H1975 cells via inhibiting miR-378a.58 The lncRNA ACTA2-AS1 suppressed malignancy by decoying miR-378a-3p, thus enhancing SOX7 expression.59 As for the downstream regulation, miR-378a works on different targets and pathways to produce a plethora of effects on lung cancer progression. Studies on stimulating miR-378a for promoting lung cancer progression and constructing miR-378a overexpression cell lines have shown that miR-378a can strengthen metastasis by regulating EMT.27 Further, miR-378a targets RBX1, and miR-378 upregulation boosts NSCLC cell invasion by downregulating RBX1, but it did not have any major effect on the angiogenesis signaling pathway. Likewise, HMOX1 was another target of miR-378a, and the interplay between HMOX1 and miR-378a29 modulated the miRNA transcriptome and the 3′ UTR of HMOX1 in tumors. Further experiments in NCI-H292 cells with stable overexpression of miR-378a indicated that miR-378a enhanced the proliferation, migration, and angiogenic capabilities of the cells both in vitro and in vivo by downregulating HMOX1 mRNA and protein expression. However, when HMOX1 was diminished, miR-378a could not modulate the cell's resistance to chemotherapeutic drugs and oxidative stress. Another study noted a reverse correlation between miR-378a and FOXG1.28 Silencing FOXG1 led to tumor suppression in NSCLC cells by downregulating miR-378a, thereby inhibiting NSCLC cell proliferation, promoting apoptosis, and increasing the length of the G0/G1 phase in the cell cycle. Hence, all the above studies show a positive relation between miR-378a and tumorigenesis.

However, some studies have shown the opposite result. Cisplatin (CDDP)-sensitive patients have high expression of miR-378a and low expression of small cell lung cancer (SCLU).25 Forced upregulation of miR-378a inhibits neoplasm growth and SCLU expression and makes cells more sensitive to CDDP. Overexpression of miR-378a-3p can inhibit cell proliferation and decrease the expression of proliferation-related proteins CDK4 and CDK6. Since lncRNA OIP5-AS1 functioned as a competing endogenous RNA of miR-378a-3p, overexpressed wild-type OIP5-AS1 increased CDK4 and CDK6 expression, thereby promoting tumor growth.60 In addition, SCP inhibits tumorigenesis by upregulating TUSC2 by targeting miR-378a-5p.61 Thus, the aberrant miR-378a expression in lung cancer significantly affects tumors. However, the controversy over whether it is the low or high expression of miR-378a that increases lung cancer progression is yet to be resolved.

Gynecological Cancer

Ovarian cancer (OC) and cervical cancer (CC) are the most common gynecological tumors. In OC and CC, miR-378a is overexpressed both in the tumor tissues and cancer cell lines.31,33 Among recurrent OC patients treated with bevacizumab, higher miR-378a correlated to longer PFS. miR-378a and its downstream targets ALCAM and EHD1 can be used as markers for anti-angiogenic therapy.62 In OC tissues and cells, it has been shown that circATRNL1 is under-expressed while miR-378a is overexpressed.32 Further study revealed that circATRNL1 could adhere to miR-378a while miR-378a directly acts on SMAD4. High circATRNL1 expression also reduces miR-378a, which, in turn, inhibits SMAD4 expression, resulting in the suppression of angiogenesis, cell proliferation, invasion, and migration, both in vitro and in vivo. Hence, in OC, circATRNL1 acts as a miR-378a sponge, thereby accelerating SMAD4 signaling and attenuating angiogenesis and metastasis.32

miR-378a is also a target of circ-LOPD2. circ-LOPD2 expression is high in both OC tissue and cells and is responsible for promoting OC cell proliferation.63 Several studies have revealed that circ-RNAs worked as molecule sponges of miR-378a to regulate cancer development.

As for CC, miR-378a acts as an oncogene since high miR-378a expression significantly enhances cancer migration and invasion in vitro, and metastasis in vivo, while downregulating miR-378a produces the opposite effect in vitro.33 The lncRNA LINC00641 inhibits CC progression by diminishing miR-378a-3p. Further, knockout of CPEB3, a downstream target of miR-378a-3p, can reverse the effects induced by LINC00641 overexpression.64 Likewise, ATG12 targets miR-378a and their expression is inversely correlated, thereby promoting tumor cell progression.33 Moreover, overexpressed miR-378a facilitates cell cycle progression, reduces apoptosis, and promotes cell growth by attenuating ST7L expression. miR-378a upregulation also activates the Wnt/β-catenin pathway to regulate tumor progression.65

However, miR-378a-3p is lower in the serum and tissues of CC patients than in the serum and tissues of healthy control subjects and normal tissues, leading to the poor prognosis of patients with CC.26 In this study, high miR-378a-3p expression reduced the proliferation and migration of CC cells both in vitro and in vivo. In short, miR-378a functions as an onco-miRNA by being regulated by circ-RNAs and then regulating its downstream targets and pathways, which is crucial in the malignancy of gynecological tumors. However, due to some inconsistent results, more studies are needed to elucidate the exact function of miR-378a in gynecological tumors.

Melanoma

Melanoma is the most aggressive type of skin cancer with high mortality.66 The miRNA miR-378a shows the highest upregulation in advanced melanoma patients and those with lymph node metastasis35 and miR-378a-5p is upregulated in metastatic melanoma patients, especially those resistant to targeted therapy. High miR-378a-5p expression enhances cell invasion, migration, and activates EMT in melanoma cells, and promotes angiogenesis. Further, several studies implementing various approaches have demonstrated that miR-378a-5p plays a carcinogenic role by regulating downstream targets such as SUFU, FUS-1, KLF9, STAMBP, and HOXD10.67 However, when miR-378a targets the 3′ UTR of FOXN3, it mitigates the stimulation of miR-378a for proliferation, migration, and invasion. Moreover, FOXN3 interacts with β-catenin to downregulate the Wnt/β-catenin signaling proteins,53 and this pathway is crucial for cell proliferation, self-regeneration, differentiation, and tissue homeostasis.68-70

Other Cancers

Apart from the positive function of miR-378a in the tumors mentioned above, similar results have been reported in nasopharyngeal carcinoma (NPC), osteosarcoma, cholangiocarcinoma, renal carcinoma, Burkitt lymphoma (BL), acute lymphoblastic leukemia (AML), and chronic lymphoblastic leukemia (CML). In NPC, miR-378a is upregulated in NPC tissues and cell lines.34 Elevating the expression of miR-378a dramatically promotes the capability of NPC to proliferate, migrate, and invade in vitro, as well as grow in vivo by restraining the expression of the TOB2 transducer. TOB2 is a potential tumor suppressor which inhibits cell proliferation by arresting the progression of the G0/G1 phase cells to the S phase.71 In osteosarcoma cells and patient-derived tumor specimens, overexpression of miR-378a promotes osteosarcoma cell proliferation by diminishing the levels of KLF9. Forced upregulation of KLF9 extraneously reverses the tumor proliferation-promoting effect of miR-378a.36 miR-378a-3p upregulation can repress cell invasion and suppress EMT by suppressing BYSL.72

Elevated miR-378a exerts a similar promoting effect in cholangiocarcinoma, which is significantly associated with an advanced TNM stage, positive lymph node metastasis, and short OS, and a miR-378a knockout inhibits cell proliferation, migration, and invasion.37 Similarly, miR-378a was identified as a diagnostic biomarker when renal carcinoma cancer tissues and cells showed increased proliferation,73-76 and increased miR-378a inhibited POLR2A and RUNX2 expression and subsequently promoted cell apoptosis.77

In BL, miR-378a-3p can facilitate tumor growth by targeting IRAK4 and MNT.39 miR-378a dysregulation is also detected in non-solid tumors, such as AML and CML. The upregulation of miR-378a is positively correlated with poor survival in AML patients.38 The 5′ flanking region of miR-378a is hypomethylated in AML. Thus, miR-378a can be reactivated by demethylation after 5-aza-dC treatment, but this is unlikely to provide helpful prognostic information in AML patients.78 Knockdown of LINC00641 inhibited cell malignancy and promoted apoptosis by downregulating miR-378a by promoting ZBTB20 expression in AML.79 Besides, lower FUS-1 expression, whose expression inversely correlates with miR-378a, is linked to the poor prognosis of AML patients.80 In CML, enhanced expression of miR-378a in leukemia cell line K562 facilitates cell proliferation, clonality, and drug resistance to 5-FU, boosts expression of stem cell self-renewal markers OCT4 and c-Myc, and suppresses apoptosis.21 As in AML, FUS-1 was also identified as a target of miR-378a in CML, but its role has not been elucidated yet.21 Together, miR-378a overexpression exerts a stimulative effect on tumors mentioned above by regulating its different targets.

Downregulation of miR-378a in Several Malignancies Suggestive of its Antitumorigenic Roles

Unlike the cancers overexpressing miR-378a, it is worth noting that miR-378a is downregulated in many cancers. An inverse relationship between miR-378a expression and tumors progression has been reported in various carcinomas, including gastric cancer (GC), colorectal cancer (CRC), liver cancer, glioma, prostate cancer (PCa), BC, MB, pituitary cancer, oral squamous carcinoma (OSCC), bladder cancer, esophageal carcinoma (ESCC), RMS, and RB.

Gastric Cancer

GC has a high incidence worldwide and severely affects the quality of life.81 Several studies have explored the underlying molecular alterations of miR-378a mediating tumorigenesis and GC development. A marked reduction of miR-378a was detected in GC tissues and cells compared to their controls.24 Underexpression of miR-378a led to poor prognosis and malignant clinicopathologic features in GC patients. Circulating miR-378a level is considered the best biomarker for GC detection since it has a higher sensitivity and specificity than that of the serum levels of CEA and CA199.82,83 Low miR-378a expression in GC patients is also associated with an advanced TNM stage, poor tumor differentiation, high lymph node metastasis rate, and poor OS and DFS. Further, GC cells are arrested in the G0/G1 or G2/M phase after miR-378 overexpression.40

miR-378a regulates various targets to exert its function as a tumor suppressor. For example, miR-378a targets BMP2, which has a high expression in GC patients with a shorter OS and DFS. Reverse-validation experiment confirmed that miR-378a overexpression inhibits invasion, migration, and EMT of GC by modulating BMP2, suggesting that components of the miR-378a/BMP2 axis might be potential therapeutic targets of GC.24 Additionally, two other targets of miR-378a in GC—MAPK1 and VEGF—are closely associated with cancer malignancy. miR-378 inhibits MAPK1 in GC.84 Depletion of MAPK1 by RNA interference in MC-803 cells reduces cell growth, increases apoptosis, and suppresses cell migration and invasion, which corresponded to the tumor-inhibiting effects caused by miR-378a restoration. Furthermore, miR-378a can directly bind to GAPLINC and reduce its expression, thereby repressing MAPK1 expression.54 Upregulated miR-378a suppresses MAPK1 expression, cell proliferation, and cell cycle progression in GC cells, while these effects are reversed by increasing the expression of the lncRNA GAPLINC. Another miR-378 target in GC is VEGF,85 as confirmed by the evidence that VEGF is downregulated by exogenously overexpressed miR-378.40 Together, these findings suggest that miR-378 regulates GC development and can act as a promising biomarker for the treatment of GC patients.

Colorectal Cancer

Decreased miR-378a expression significantly correlates with shorter OS in CRC patients.23,86 Hence, miR-378a is a potential biomarker for the early detection and diagnosis of patients with CRC.23,87,88 Interestingly, miR-378a might also function as a novel biomarker to predict the efficacy of vaccines against CRC.89,90 The overexpression of miR-378a in CRC repressed cell growth,23 induced apoptosis,86 and inhibited migration and invasion by restraining EMT.55 Studies also showed that miR-378a mitigated the malignant phenotypes of CRC cells by inhibiting the Wnt/ β-catenin pathway.91

Two lncRNAs were found to regulate miR-378a-3p. SP1-activated LINC00339 decreased miR-378a-3p but enhanced MED19 expression, contributing to cell proliferation, cell cycle progression, migration, and invasion in CRC cells.91 Further, the lncRNA CYTOR decreased miR-378a-5p and led to the increase in SERPINE1, which inhibited L-OHP resistance and EMT.92

Several other genes are the downstream targets of miR-378a, including IGF1R, CDC40, vimentin, BRAF, SDAD1, and KISS1. IGF1R protein expression significantly negatively correlated with miR-378a in CRC tissues.86 Further, luciferase reporter assays confirmed CDC40 as a direct target of miR-378a.41 miR-378a represses cell growth and G1/S transition in CRC cells, and consistent with the inhibitory effect of miR-378a on tumor growth, CDC40 knockdown reduces proliferation and represses cell cycle progression by regulating G1/S and G2/M phases and pre-mRNA splicing.

High miR-378a expression restrains CRC cell growth and invasion and downregulates vimentin while silencing miR-378a produces the inverse effects.23 miR-378 diametrically targets the 3′ UTR of vimentin to inhibit tumor progression. Similarly, BRAF55 and SDAD193 were significantly reduced by miR-378a-5p overexpression in CRC. SDAD1 expression increased upon blocking miR-378a, suggesting that miR-378a and SDAD1 have a direct but negative regulatory relationship. Further, the expression of KiSS1 is enhanced in CRC cells with miR-378a elevation, leading to decreased proliferation, migration, and invasion abilities of the cancer cells.94

Liver Cancer

Low expression of miR-378a95-97 is related to a poor prognosis of liver cancer.97 Enhanced miR-378a expression inhibits the proliferation and invasion98 but increases the apoptosis99 in liver cancer. Moreover, miR-378a elevated the sensitivity of sorafenib treatment by targeting VEGFR, PDGFRβ, and c-Raf.98

miR-378a also enhances hepatocellular carcinoma (HCC) development by downregulating FUS expression,100 while miR-378a-5p targets VEGF.101 Moreover, miR-378a targets TRAF1 and weakens NF-κB signaling, consequently downregulating VEGF.97 IGF1R was also identified as a novel target of miR-378a in sorafenib-resistant HCC cells, with high miR-378a expression activating the PI3K/AKT and the Ras/Raf/MAP kinase pathways to mediate cell survival102 and IGF1R knockout significantly restrained the miR-378a-mediated sorafenib resistance. miR-378a also suppresses HCC tumorigenesis by downregulating PD-L1 and STAT3 expression.103 Moreover, the overexpressed miR-378a suppressed HCC cell proliferation by arresting G2/M and inhibited tumor growth in vivo.42 Interestingly, in HCC, circCRIM1 acts as an upstream regulator of miR-378a that upregulates SKP2 by acting as a sponge for miR-378a and facilitates cell proliferation, angiogenesis, and the transition from the G1 to the S phase in the cell cycle.104

Glioma

Even with the advancement of therapeutic strategies, glioma still shows a poor prognosis due to its high tumor aggressiveness and unlimited proliferation.105,106 Serval clinical studies in glioma have shown that miR-378a is remarkably decreased in cancer tissue and can potentially act as a diagnostic biomarker.107-109 Glioma patients with lower miR-378a expression showed poorer OS, and miR-378a overexpression restrained cell migration and invasion.109 Downregulated miR-378a increased the target IRG1 expression, which enhanced glioma cell growth, invasion, migration, and EMT.108 Another study showed that TSPAN17 was a direct target of miR-378a and correlated with poor prognosis in glioblastoma patients. Overexpressed miR-378a weakened TSPAN17 expression, consequently promoting apoptosis and reducing proliferation, migration, and invasion. These effects were attenuated in rescue experiments increasing TSPAN17 expression.110 Interestingly, interfilamentous vimentin can also mediate miR-378a self-renewal by regulating SOX2 transcription factor expression.111 Further, miR-378a promotes cell survival, tumor growth, and angiogenesis by targeting SUFU and FUS-1 expression.112

Prostate Cancer

miR-378a is notably underexpressed in PCa tissues.44 The reduction of miR-378a levels resulted in higher Gleason score, larger diameter tumors, and elevated serum prostate-specific antigen in PCa.113,114 miR-378a improved risk stratification based on the Gleason score or tumor stage and led to a higher risk of recurrence in patients with low miR-378a levels. Thus, miR-378a is a promising independent predictor of short-term recurrence in patients at high and very high risk of recurrence.113

CircRNA PDHX boosts PCa progression by sponging miR-378a.115 MiR-378a also inhibits prostate cell proliferation and glucose metabolism by repressing GLUT1.114 Forced miR-378a overexpression represses prostate cancer cell migration and invasion but promotes cell apoptosis in vitro.44 Further, studies have shown that KLK2, KLK4,113 and MAPK1 are the targets of miR-378a in PCa, and the ectopic expression of MAPK1 rescues miR-378a-inhibited cell migration and invasion capacity.44

Breast Cancer

Even in BC, miR-378a shows a low expression and correlates with the unfavorable prognosis of BC patients upon tamoxifen treatment.116 GOLT1A is regulated by miR-378a and is critical for the mechanisms underlying BC endocrine resistance.116 The m6A-mediated increase in LINC00958 expression enhanced tumorigenesis through the miR-378a/YY1 pathway.117 Further, DCTPP1 is modulated by miR-378a, and decreased miR-378a expression elevates the DCTPP1 level, which facilitates cell proliferation by activating DNA repair signaling.118

RUNX1 is significantly increased upon inhibition of miR-378a expression, which also enhances the invasion and migration of the triple-negative breast cancer (TNBC) cells. Further, the regulation of RUNX1 is mediated by the PPARGC1Β/miR-378a/RUNX1 regulatory pathway.45 It has also been revealed that lncRNA GAS5 and miR-378a bind to each other, and the target of miR-378a-5p, SUFU, promotes GAS5-induced apoptosis of TNBC cells. This implicates that GAS5 stimulates apoptosis in TNBC cells by regulating miR-378a-5p/SUFU signaling.119 Moreover, although no significant effect of miR-378a was found on KLK4 expression, both miR-378a and KLK4 represent unfavorable prognostic markers in TNBC patients.120

Other Cancers

In addition to the above-mentioned tumors, decreased miR-378a expression has also been reported in other cancers. miR-378a is significantly reduced in MB. miR-378a negatively regulates UHRF1 expression in MB by binding to the 3′ UTR of its transcript.46 Therefore, miR-378a overexpression inhibits UHRF1, but increased expression of UHRF1 reverses the miR-378a-induced suppression of cell proliferation and promotion of apoptosis.

In PA, RNF31 is highly expressed, and its expression is negatively regulated by miR-378a.47 Knocking out RNF31 inhibits the proliferation and migration of the pituitary tumor cell line GH3. Similarly, miR-378a plays an inhibitory role in oral squamous cell carcinoma (OSCC). miR-378a is downregulated121 in OSCC and also targets KLK4.48 Overexpressed KLK4 reverses the miR-378a-induced suppression of migration and invasion by enhancing MMP-9, MMP-2, and N-cadherin expression while decreasing E-cadherin levels. CircRNA-100290 can also rescue the miR-378a-induced suppression of GLUT1 by functioning as a competing endogenous RNA (ceRNA), thereby accelerating cell proliferation and glycolysis in OSCC.122 NCAPG, a target of miR-378a in OSCC, promotes cell proliferation and cell cycle progression but inhibits apoptosis by activating the GSK-3β/β-catenin signaling in OSCC.123

In bladder cancer, miR-378a is downregulated and can act as an independent prognostic factor for the OS and recurrence.49 LINC00958, an upstream regulator of miR-378, promotes the proliferation and metastasis of bladder cancer cells by increasing IGF1R, a downstream target of miR-378a, by sponging miR-378a.124 miR-378a is lower in esophageal squamous cell carcinoma (ESCC) cells and tissues than in normal cells and adjacent tissues. Low miR-378a expression leads to an unfavorable prognosis and shorter OS in ESCC patients. Further, low miR-378a levels elevate cell proliferation, migration, and invasion.50 However, miR-378a overexpression exerts the opposite effect by inhibiting Rab10.125 Mechanistically, the lncRNAs SLC2A1-AS1 and LINC00514 are essential in ESCC progression by acting as miR-378a sponges. SLC2A1-AS1 inhibits miR-378a-5p by regulating glycolysis, thereby restoring GLUT1 expression and enhancing ESCC.126 LINC00514, acting as a ceRNA, upregulates SPHK1 by sponging miR-378a-5p and activates adipogenesis-related pathways, thereby promoting the proliferation and invasion of ESCC cells.127

In a RMS-derived cell line, miR-378a upregulation inhibited IGF1R expression and impacted phosphorylated-Akt protein levels.51 Moreover, ectopic expression of miR-378a modulated apoptosis, cell migration, cytoskeleton organization, and the expression of the muscle markers MyoD1, MyoR, desmin, and MyHC in RMS. DNA demethylation by 5-aza-2 -deoxycytidine (5-aza-dC) also elevates miR-378a levels which correlate with increased apoptosis, cell viability reduction, and cell cycle arrest in G2-phase.51 In RB, upregulated miR-378a-3p inhibits cell proliferation by targeting FOXG1.52

The Role of miR-378a in Anti-Cancer Therapeutic Resistance

Unfortunately, cancer cells cumulatively get resistant to miscellaneous anti-tumor therapies, leading to disease recurrence and metastasis. Hence, understanding the internal molecular mechanism of cancer drug resistance is essential for finding new therapeutic approaches. Given that miR-378 is involved in various biological processes, the abnormal expression of miR-378a might be responsible for the anti-cancer resistance. For instance, miR-378a expression is preternatural in chemotherapy resistance in esophageal cancer cells.128 Many studies have also shown miR-378a as a promising candidate for anti-tumor treatment. In lung cancer, elevated miR-378a recovered CDDP chemosensitivity by targeting SCLU and downregulating Bcl-2, pCas-3, p-Erk1/2, and p-Akt.25 In ovarian cancer, low miR-378a expression can elevate the efficacy of bevacizumab treatment in patients with recurrent ovarian cancer, thereby lengthening PFS. Further, multivariate analysis showed miR-378a level as an efficient predictor of PFS after anti-angiogenic therapy.62

In CRC cells with upregulated miR-378a, sensitivity to cetuximab was restored in all BRAF mutants and half of KRAS mutants, and lauric acid improved the sensitization of Cetuximab in KRAS/BRAF mutated CRC cells by regaining miR-378a expression.129 Moreover, EPA-induced upregulation of miR-378 led to the significant restoration of sensitivity to cetuximab in the KRAS-mutant cells.130In glioma, the inhibitory effect of curcumin was enhanced in miR-378a-expressing stable U87 cells.43 Likewise, increased miR-378a expression enhances radiation response by promoting radiation-induced TGD in glioblastoma cells.107 However, reduced miR-378a expression was found in sorafenib-resistant HCC cells than in the sorafenib-sensitive group. Further, LXR stimulated miRNA-378a-3p transcription and hence can be used as a potential combinable treatment strategy with sorafenib to suppress HCC progression.102 Metformin also induces miR-378a to downregulate CDK1, leading to suppression of cell proliferation and induction of the G2/M cell cycle arrest in HCC.42 Moreover, the biologically active component isolated from the fungus Ganoderma lucidum can also overcome the drug resistance conferred by miR-378.131

Conclusion

Cancer is the leading cause of death worldwide. The high mortality rate of cancers can be ascribed to its inefficient early detection, inherent or acquired therapeutic resistance, and poor predictive power of conventional screening techniques and clinicopathological parameters. Hence, searching for novel molecular targets is necessary to solve this problem.

The miRNA miR-378a is a promising biomarker and therapeutic target for cancers. As discussed in this paper, miR-378a expression is aberrant in more than 23 cancers and is responsible for affecting the cell's capability to proliferate, invade, metastasize, and resist multiple anti-tumor treatment regimens. However, there is still some controversy and ambiguity over its expression and roles in some cancers. miR-378a plays a dual role in tumors and can produce different impacts on different tumors based on whether it is overexpressed or under-expressed. The fact that it acts as an onco-miRNA makes it an attractive therapeutic target in lung cancer,22,25,27-29 CC,33 OC,30-32 NPC,34 melanoma,35 osteosarcoma,36 cholangiocarcinoma,37 AML,38 CML,21 renal carcinoma,73-76 and BL.39 However, it acts as a tumor suppressor in GC,84 CRC ,41 liver cancer,42 glioma,43 PCa,44 BC,45 MB,46 PA,47 OSCC,48 bladder cancer,49 ESCC,50 RMS,51 and RB.52 Notably, the expression and role of miR-378a are still controversial in malignant tumors, including lung cancer60 and CC.26 All these effects may be due to the different expression of the two strands of miR-378a and it is also due to the different specific expression of miR-378a in tissues and cells of different tumors. Further, the differences and limitations in the source of cell and tissue samples might be the key influencing factors.

The expression of circulating miR-378a can be used for cancer screening and diagnosis.22-24,31,33 The identification of the signaling pathways involved in cell proliferation,34 migration,35 invasion,33 and apoptosis41 also provides evidence for the different functions of miR-378a. miR-378a has also shown value as a therapy sensitizer25 and drug target.25,131 All the results mentioned above might be due to the regulation of miR-378a by various factors, such as circRNA and lncRNA, and the interaction between miR-378a and its targets that constitute complex regulatory networks. Depending on the cellular environment, miR-378a might have various biological functions and clinical applications and can even be expressed differently and have varied roles in different tumors.

Because of the dual function of miR-378a in cancer development and whether targeting miR-378a is an effective and feasible approach for individual tumor patient-based treatment, more detailed investigations and understanding of the mechanisms underlying how miR-378a regulates tumor progression and therapy resistance are required in the future. Different analysis methods and intervention strategies for the role of the miRNA in different cancers are essential. Further, considering that miR-378a can effectively regulate the response of cancer cells to chemotherapy25,128 and radiotherapy,43 a combination of miR-378a targeted therapy and chemotherapy or radiotherapy might achieve better therapeutic effects in some cancer types.

Thus, in this review, we provide insights into the rationale underlying the dual functions of miRNAs in tumor onset and progression. We show that miR-378 is critical for key biological and pathological processes via its complex network regulation mechanism in human cancers. In the future, miR-378 has great potential to become a clinically effective approach for cancer diagnosis and prognosis by refining cancer types and subtypes and an effective therapeutic strategy against cancer.

Abbreviations

ALCAM

activated leukocyte cell adhesion molecule

AML

acute myelocytic leukemia

ATG12

autophagy-related protein 12

BC

breast cancer

BL

Burkitt lymphoma

BM

brain metastasis

BMP2

bone morphogenetic proteins 2

BMNCs

bone marrow mononuclear cells

BRAF

B-Raf proto-oncogene serine/threonine kinase

BYSL

bystin like

CC

cervical cancer

CDC40

cell division cycle 40

CDDP

cisplatin

CDK

cyclin dependent kinase

CML

chronic myeloid leukemia

CRC

colorectal cancer

DCTPP1

dCTP pyrophosphatase 1

DFS

disease-free survival

EMT

epithelial-mesenchymal transition

EHD1

EH domain containing 1

EPA

eicosapentaenoic acid ethyl ester

ESCC

esophageal carcinoma

FOXG1

Fork head-box gene 1

FOXN3

Forkhead box N3

FUS-1

FUS RNA binding protein

FOXN3

forkhead box N3

GC

gastric cancer

GLUT1

glucose transporter 1

GOLT1A

golgi Transport 1A

HCC

hepatocellular carcinoma

HMOX1

heme oxygenase-1

HOXD10

homeobox D10

IRAK4

interleukin-1 receptor associated kinase-4

IRG1

immune responsive gene 1

KLF9

Fruppel-like factor 9

KLK

Kallikrein-related peptidase

KISS1

tumor metastasis suppressor kiss-1

LXR

liver X receptor

miR-378a

MicroRNA-378a

MAPK1

mitogen-activated protein kinase 1

MB

medulloblastoma

MED19

mediator complex subunit 19

MNT

MAX network transcriptional repressor

NPC

nasopharyngeal carcinoma

NSCL

non-small cell lung carcinoma

OC

ovarian cancer

OCT4

organic cation/carnitine transporter 4

OS

overall survival

OSCC

oral squamous carcinoma

PA

pituitary adenoma

PCa

prostate cancer

PDGFRβ

platelet derived growth factor receptor beta

PFS

progression-free survival

POLR2A

RNA polymerase II subunit A

Ppargc1β

peroxisome proliferator-activated receptor γ coactivator 1 beta

PSA

prostate-specific antigen

RB

retinoblastoma

RBX1

ring-box 1

RMS

rhabdomyosarcoma

RNF31

ring finger protein 31

RUNX1

runt related transcription factor 1

SCLU

secreted form clusterin

SDAD1

SDA1 domain containing 1

SERPINE1

serpin family E member 1

SKP2

S-phase kinase associated protein 2

SMAD4

SMAD family member 4

SOX2

SRY-box transcription factor 2

SOX7

SRY-box transcription factor 7

STAMBP

STAM binding protein

ST7L

suppression of tumorigenicity 7 like

STAT3

signal transducer and activator of transcription 3

SUFU

SUFU negative regulator of hedgehog signaling

TOB2

transducer of ERBB2

TSPAN17

tetraspanin 17

TNBC

triple-negative breast cancer

TUSC2

tumor suppressor 2, mitochondrial calcium regulator

UHRF1

ubiquitin like with PHD and ring finger domains 1

VEGF

vascular endothelial growth factor

ZBTB20

zinc finger and BTB domain containing 20

Footnotes

Author Contributions: YQ and RL conceived the idea, YQ and LL performed literature retrieval, YQ conducted literature reading and sorting, YQ wrote initial article and revised the manuscript, RL provided helpful ideas and comments on this article, PL played an essential role in manuscript revision and responding to comments, XZ supervised the study. All the authors have read and approved the final version of this article. YQ and RL have contributed equally to this work as co-first author.

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was supported by The Independent Project of Key Laboratory of Early Prevention and Treatment for Regional High-Incidence Tumor (grant number GKE-22202120).

References

  • 1.Hutvagner G, Zamore PD. A microRNA in a multiple-turnover RNAi enzyme complex. Science. 2002;297(5589): 2056-2060. [DOI] [PubMed] [Google Scholar]
  • 2.He L, Hannon GJ. MicroRNAs: Small RNAs with a big role in gene regulation. Nat Rev Genet. 2004;5(7):522-531. [DOI] [PubMed] [Google Scholar]
  • 3.Rosalind C, Lee RLF, Ambrost V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-4.pdf. Cell. 1993;75(5):843-854. [DOI] [PubMed] [Google Scholar]
  • 4.Shukla V, Varghese V, Kabekkodu S, et al. Enumeration of deregulated miRNAs in liquid and tissue biopsies of cervical cancer. Gynecol Oncol. 2019;155(1):135-143. [DOI] [PubMed] [Google Scholar]
  • 5.Wang CH, Lee DY, Deng Z, et al. MicroRNA miR-328 regulates zonation morphogenesis by targeting CD44 expression. PLoS One. 2008;3(6):e2420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kawasaki H, Taira K. Retraction: Hes1 is a target of microRNA-23 during retinoic-acid-induced neuronal differentiation of NT2 cells. Nature. 2003;426(6962): 100. [DOI] [PubMed] [Google Scholar]
  • 7.Chan JA, Krichevsky AM, Kosik KS. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005;65(14):6029-6033. [DOI] [PubMed] [Google Scholar]
  • 8.Xu C LY, Pan Z, Chu W, et al. The muscle-specific microRNAs miR-1 and miR-133 produce opposing effects on apoptosis by targeting HSP60, HSP70 and caspase-9 in cardiomyocytes. J Cell Sci. 2011;124(Pt 18):3187. [DOI] [PubMed] [Google Scholar]
  • 9.Shatseva T, Lee DY, Deng Z, Yang BB. MicroRNA miR-199a-3p regulates cell proliferation and survival by targeting caveolin-2. J Cell Sci. 2011;124(Pt 16):2826-2836. [DOI] [PubMed] [Google Scholar]
  • 10.Croce CM, Calin GA. miRNAs, cancer, and stem cell division. Cell. 2005;122(1):6-7. [DOI] [PubMed] [Google Scholar]
  • 11.Mello C, Czech M. Micromanaging insulin secretion. Nat Med. 2004;10(12):1297-1298. [DOI] [PubMed] [Google Scholar]
  • 12.Chellappan P, Vanitharani R, Fauquet C. MicroRNA-binding viral protein interferes with Arabidopsis development. Proc Natl Acad Sci U S A. 2005;102(29):10381-10386. doi: 10.1073/pnas.0504439102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Iorio M, Croce C. Causes and consequences of microRNA dysregulation. Cancer journal (Sudbury). Mass. 2012;18(3):215-222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Visone R, Croce C. MiRNAs and cancer. Am J Pathol. 2009;174(4):1131-1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wu W, Lee C, Cho C, et al. MicroRNA dysregulation in gastric cancer: A new player enters the game. Oncogene. 2010;29(43):5761-5771. [DOI] [PubMed] [Google Scholar]
  • 16.Labbaye C, Testa U. The emerging role of MIR-146A in the control of hematopoiesis, immune function and cancer. J Hematol Oncol. 2012;5:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kasashima K, Nakamura Y, Kozu T. Altered expression profiles of microRNAs during TPA-induced differentiation of HL-60 cells. Biochem Biophys Res Commun. 2004;322(2):403-410. [DOI] [PubMed] [Google Scholar]
  • 18.Carrer M, Liu N, Grueter C, et al. Control of mitochondrial metabolism and systemic energy homeostasis by microRNAs 378 and 378*. Proc Natl Acad Sci U S A. 2012;109(38):15330-15335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Eichner L, Perry M, Dufour C, et al. miR-378(∗) mediates metabolic shift in breast cancer cells via the PGC-1β/ERRγ transcriptional pathway. Cell Metab. 2010;12(4):352-361. [DOI] [PubMed] [Google Scholar]
  • 20.Yu J, Kong X, Liu J, et al. Expression profiling of PPARγ-regulated microRNAs in human subcutaneous and visceral adipogenesis in both genders. Endocrinology. 2014;155(6):2155-2165. [DOI] [PubMed] [Google Scholar]
  • 21.Ma J, Wu D, Yi J, et al. MiR-378 promoted cell proliferation and inhibited apoptosis by enhanced stem cell properties in chronic myeloid leukemia K562 cells. Biomedicine Pharmacotherapy = Biomedecine Pharmacotherapie. 2019;112:108623. [DOI] [PubMed] [Google Scholar]
  • 22.Chen L, Xu S, Xu H, Zhang J, Ning J, Wang S. MicroRNA-378 is associated with non-small cell lung cancer brain metastasis by promoting cell migration, invasion and tumor angiogenesis. Med Oncol (Northwood, London, England). 2012;29(3):1673-1680. [DOI] [PubMed] [Google Scholar]
  • 23.Zhang G, Zhou H, Xiao H, Li Y, Zhou T. MiR-378 is an independent prognostic factor and inhibits cell growth and invasion in colorectal cancer. BMC Cancer. 2014;14:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yang Y, Luo S, Wang L. Effects of microRNA-378 on epithelial-mesenchymal transition, migration, invasion and prognosis in gastric carcinoma by targeting BMP2. Eur Rev Med Pharmacol Sci. 2019;23(12):5176-5186. [DOI] [PubMed] [Google Scholar]
  • 25.Chen X, Jiang Y, Huang Z, et al. miRNA-378 reverses chemoresistance to cisplatin in lung adenocarcinoma cells by targeting secreted clusterin. Sci Rep. 2016;6:19455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Zhang L, Wu Z. MicroRNA-378a-3p downregulation as a novel biomarker with poor clinical outcomes in cervical cancer. Biomed Environ Sci: BES. 2021;34(3):213-221. [DOI] [PubMed] [Google Scholar]
  • 27.Ho C, Noor S, Nagoor N. RBX1MiR-378 And MiR-1827 regulate tumor invasion, migration and angiogenesis in human lung adenocarcinoma by targeting and, respectively. J Cancer. 2018;9(2):331-345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ji K, Cui F, Qu D, et al. MiR-378 promotes the cell proliferation of non-small cell lung cancer by inhibiting FOXG1. Eur Rev Med Pharmacol Sci. 2018;22(4):1011-1019. [DOI] [PubMed] [Google Scholar]
  • 29.Skrzypek K, Tertil M, Golda S, et al. Interplay between heme oxygenase-1 and miR-378 affects non-small cell lung carcinoma growth, vascularization, and metastasis. Antioxid Redox Signaling. 2013;19(7):644-660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Heldin C, Moustakas A. Role of Smads in TGFβ signaling. Cell Tissue Res. 2012;347(1):21-36. [DOI] [PubMed] [Google Scholar]
  • 31.Kim Y, Kim E, Jeon D, et al. Differential microRNA expression signatures and cell type-specific association with Taxol resistance in ovarian cancer cells. Drug Des Devel Ther. 2014;8:293-314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wang J, Li Y, Zhou J, Shen F, Shi X, Chen Y. CircATRNL1 activates Smad4 signaling to inhibit angiogenesis and ovarian cancer metastasis via miR-378. Mol Oncol. 2021;15(4):1217-1233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Tan D, Zhou C, Han S, Hou X, Kang S, Zhang Y. MicroRNA-378 enhances migration and invasion in cervical cancer by directly targeting autophagy-related protein 12. Mol Med Rep. 2018;17(5):6319-6326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yu B, Peng X, Zhao F, et al. MicroRNA-378 functions as an onco-miR in nasopharyngeal carcinoma by repressing TOB2 expression. Int J Oncol. 2014;44(4):1215-1222. [DOI] [PubMed] [Google Scholar]
  • 35.Sun M, Ma X, Tu C, et al. MicroRNA-378 regulates epithelial-mesenchymal transition and metastasis of melanoma by inhibiting FOXN3 expression through the Wnt/β-catenin pathway. Cell Biol Int. 2019;43(10):1113-1124. [DOI] [PubMed] [Google Scholar]
  • 36.Peng N, Miao Z, Wang L, Liu B, Wang G, Guo X. MiR-378 promotes the cell proliferation of osteosarcoma through down-regulating the expression of Kruppel-like factor 9. Biochem Cell Biol = Biochimie Biologie Cellulaire. 2018;96(5):515-521. [DOI] [PubMed] [Google Scholar]
  • 37.Zhou Z, Ma J. miR-378 serves as a prognostic biomarker in cholangiocarcinoma and promotes tumor proliferation, migration, and invasion. Cancer Biomarkers: Section A of Disease Markers. 2019;24(2):173-181. [DOI] [PubMed] [Google Scholar]
  • 38.Qian J, Lin J, Qian W, et al. Overexpression of miR-378 is frequent and may affect treatment outcomes in patients with acute myeloid leukemia. Leuk Res. 2013;37(7):765-768. [DOI] [PubMed] [Google Scholar]
  • 39.Niu F, Dzikiewicz-Krawczyk A, Koerts J, et al. MiR-378a-3p Is critical for Burkitt lymphoma cell growth. Cancers. 2020;12(12):3546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Deng H, Guo Y, Song H, et al. MicroRNA-195 and microRNA-378 mediate tumor growth suppression by epigenetical regulation in gastric cancer. Gene. 2013;518(2):351-359. [DOI] [PubMed] [Google Scholar]
  • 41.Wang K, Ma J, Zhang F, Yu M, Xue J, Zhao J. MicroRNA-378 inhibits cell growth and enhances L-OHP-induced apoptosis in human colorectal cancer. IUBMB life. 2014;66(9):645-654. [DOI] [PubMed] [Google Scholar]
  • 42.Zhou J, Han S, Qian W, Gu Y, Li X, Yang K. Metformin induces miR-378 to downregulate the CDK1, leading to suppression of cell proliferation in hepatocellular carcinoma. Onco Targets Ther. 2018;11:4451-4459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Li W, Yang W, Liu Y, et al. MicroRNA-378 enhances inhibitory effect of curcumin on glioblastoma. Oncotarget. 2017;8(43):73938-73946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Chen Q, Zhou W, Han T, et al. MiR-378 suppresses prostate cancer cell growth through downregulation of MAPK1 in vitro and in vivo. Tumour Biol: J Int Soc Oncodevelop Biol Med. 2016;37(2):2095-2103. [DOI] [PubMed] [Google Scholar]
  • 45.Browne G, Dragon J, Hong D, et al. MicroRNA-378-mediated suppression of Runx1 alleviates the aggressive phenotype of triple-negative MDA-MB-231 human breast cancer cells. Tumour Biol: J Int Soc Oncodevelop Biol Med. 2016;37(7):8825-8839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zhang Z, Zhu B, Zhao X, et al. Regulation of UHRF1 by microRNA-378 modulates medulloblastoma cell proliferation and apoptosis. Oncol Rep. 2017;38(5):3078-3084. [DOI] [PubMed] [Google Scholar]
  • 47.Qiu P, Xu T, Lu X, Yang W, Zhang Y, Xu G. MicroRNA-378 regulates cell proliferation and migration by repressing RNF31 in pituitary adenoma. Oncol Lett. 2018;15(1):789-794. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 48.Cui Z, Sun S, Liu Q, et al. MicroRNA-378-3p/5p suppresses the migration and invasiveness of oral squamous carcinoma cells by inhibiting KLK4 expression. Biochem Cell Biol = Biochimie Biologie Cellulaire. 2020;98(2):154-163. [DOI] [PubMed] [Google Scholar]
  • 49.Zaravinos A, Radojicic J, Lambrou G, et al. Expression of miRNAs involved in angiogenesis, tumor cell proliferation, tumor suppressor inhibition, epithelial-mesenchymal transition and activation of metastasis in bladder cancer. J Urol. 2012;188(2):615-623. [DOI] [PubMed] [Google Scholar]
  • 50.Jin W, Wang L, Cheng S, Lv H. Prognostic value of microRNA-378 in esophageal cancer and its regulatory effect on tumor progression. Exp Ther Med. 2021;22(1):704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Megiorni F, Cialfi S, McDowell H, et al. Deep Sequencing the microRNA profile in rhabdomyosarcoma reveals down-regulation of miR-378 family members. BMC Cancer. 2014;14:880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhang C, Wu S. microRNA − 378a-3p restrains the proliferation of retinoblastoma cells but promotes apoptosis of retinoblastoma cells via inhibition of FOXG1. Invest Ophthalmol Visual Sci. 2020;61(5):31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Dai Y, Wang M, Wu H, Xiao M, Liu H, Zhang D. Loss of FOXN3 in colon cancer activates beta-catenin/TCF signaling and promotes the growth and migration of cancer cells. Oncotarget. 2017;8(6):9783-9793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Diao L, Wang S, Sun Z. Long noncoding RNA GAPLINC promotes gastric cancer cell proliferation by acting as a molecular sponge of miR-378 to modulate MAPK1 expression. Onco Targets Ther. 2018;11:2797-2804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wang Z, Ma B, Ji X, et al. MicroRNA-378-5p suppresses cell proliferation and induces apoptosis in colorectal cancer cells by targeting BRAF. Cancer Cell Int. 2015;15:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Li R, Liu Y, Wang T, et al. The characteristics of lung cancer in Xuanwei County: A review of differentially expressed genes and noncoding RNAs on cell proliferation and migration. Biomedicine Pharmacotherapy = Biomedecine Pharmacotherapie. 2019;119:109312. [DOI] [PubMed] [Google Scholar]
  • 57.Zhang Y, Xu H. Serum exosomal miR-378 upregulation is associated with poor prognosis in non-small-cell lung cancer patients. J Clin Lab Anal. 2020;34(6):e23237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gao S, Yu Y, Liu L, Meng J, Li G. Circular RNA hsa_circ_0007059 restrains proliferation and epithelial-mesenchymal transition in lung cancer cells via inhibiting microRNA-378. Life Sci. 2019;233:116692. [DOI] [PubMed] [Google Scholar]
  • 59.Ying K, Wang L, Long G, Lian C, Chen Z, Lin W. ACTA2-AS1 Suppresses lung adenocarcinoma progression via sequestering miR-378a-3p and miR-4428 to elevate SOX7 expression. Cell Biol Int. 2020;44(12):2438-2449. [DOI] [PubMed] [Google Scholar]
  • 60.Wang M, Sun X, Yang Y, Jiao W. Long non-coding RNA OIP5-AS1 promotes proliferation of lung cancer cells and leads to poor prognosis by targeting miR-378a-3p. Thorac Cancer. 2018;9(8):939-949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Mao J, Zhang Z, Chen Y, et al. Sea cucumber peptides inhibit the malignancy of NSCLC by regulating miR-378a-5p targeted TUSC2. Food Funct. 2021;12(24):12362-12371. [DOI] [PubMed] [Google Scholar]
  • 62.Chan J, Kiet T, Blansit K, et al. MiR-378 as a biomarker for response to anti-angiogenic treatment in ovarian cancer. Gynecol Oncol. 2014;133(3):568-574. [DOI] [PubMed] [Google Scholar]
  • 63.Wei X, Lv H, Yang S, Yang X. CircRNA PLOD2 enhances ovarian cancer propagation by controlling miR-378. Saudi J Biol Sci. 2021;28(11):6260-6265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zhang Y, Yu R, Li L. LINC00641 Hinders the progression of cervical cancer by targeting miR-378a-3p/CPEB3. J Gene Med. 2020;22(9):e3212. [DOI] [PubMed] [Google Scholar]
  • 65.Li S, Yang F, Wang M, Cao W, Yang Z. miR-378 functions as an onco-miRNA by targeting the ST7L/Wnt/β-catenin pathway in cervical cancer. Int J Mol Med. 2017;40(4):1047-1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Siegel R, Miller K, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67(1):7-30. [DOI] [PubMed] [Google Scholar]
  • 67.Tupone M, D’Aguanno S, Di Martile M, et al. microRNA-378a-5p iS a novel positive regulator of melanoma progression. Oncogenesis. 2020;9(2):22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Huo J, Zhang Y, Li R, Wang Y, Wu J, Zhang D. Upregulated MicroRNA-25 mediates the migration of Melanoma cells by targeting DKK3 through the WNT/β-Catenin pathway. Int J Mol Sci. 2016;17(11):1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kovacs D, Migliano E, Muscardin L, et al. The role of Wnt/β-catenin signaling pathway in melanoma epithelial-to-mesenchymal-like switching: Evidences from patients-derived cell lines. Oncotarget. 2016;7(28):43295-43314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wang E, Wang D, Li B, et al. Capn4 promotes epithelial-mesenchymal transition in human melanoma cells through activation of the Wnt/β-catenin pathway. Oncol Rep. 2017;37(1):379-387. [DOI] [PubMed] [Google Scholar]
  • 71.Winkler G. The mammalian anti-proliferative BTG/Tob protein family. J Cell Physiol. 2010;222(1):66-72. [DOI] [PubMed] [Google Scholar]
  • 72.Zhang J, Tang H, Jiang X, Huang N. Wei Q. Hypoxia-induced miR-378a-3p Inhibits osteosarcoma invasion and epithelial-to-mesenchymal via BYSL transition regulation. Front Genet. 2021;12:804952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Fedorko M, Stanik M, Iliev R, et al. Combination of MiR-378 and MiR-210 serum levels enables sensitive detection of renal cell carcinoma. Int J Mol Sci. 2015;16(10):23382-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hauser S, Wulfken L, Holdenrieder S, et al. Analysis of serum microRNAs (miR-26a-2*. miR. 191, miR-337-3p and miR-378) as potential biomarkers in renal cell carcinoma. Cancer Epidemiol. 2012;36(4):391-3914. [DOI] [PubMed] [Google Scholar]
  • 75.Redova M, Poprach A, Nekvindova J, et al. Circulating miR-378 and miR-451 in serum are potential biomarkers for renal cell carcinoma. J Transl Med. 2012;10:55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Li H, Li J, Wang Z, et al. Identification of angiogenesis-related miRNAs in a population of patients with renal clear cell carcinoma. Oncol Rep. 2014;32(5):2061-2069. [DOI] [PubMed] [Google Scholar]
  • 77.Weng W, Yu K, Jhan J, Pang S, Chiou Y, Pang S. Micro-RNA378a-3p induces apoptosis in sarcomatoid renal cell carcinoma and regulates POLR2A and RUNX2 expression. Anticancer Res. 2022;42(2):811-825. [DOI] [PubMed] [Google Scholar]
  • 78.Zhu X, Wen X, Zhang Y, et al. The 5′ flanking region of miR-378 is hypomethylated in acute myeloid leukemia. Int J Clin Exp Pathol. 2015;8(5):4321-4331. [PMC free article] [PubMed] [Google Scholar]
  • 79.Wang J, Liu Z, Yu L. Long non-coding RNA LINC00641 promotes cell growth and migration through modulating miR-378a/ZBTB20 axis in acute myeloid leukemia. Eur Rev Med Pharmacol Sci. 2019;23(17):7498-7509. [DOI] [PubMed] [Google Scholar]
  • 80.Tang X, Zhou J, Zhang J, et al. Low expression of FUS1 is negatively correlated with miR-378 and may predict adverse prognoses in acute myeloid leukemia. Acta Haematol. 2018;139(2):89-95. [DOI] [PubMed] [Google Scholar]
  • 81.Lordick F, Janjigian Y. Clinical impact of tumour biology in the management of gastroesophageal cancer. Nature Rev Clin Oncol. 2016;13(6):348-360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Guo J, Miao Y, Xiao B, et al. Differential expression of microRNA species in human gastric cancer versus non-tumorous tissues. J Gastroenterol Hepatol. 2009;24(4):652-657. [DOI] [PubMed] [Google Scholar]
  • 83.Liu H, Zhu L, Liu B, et al. Genome-wide microRNA profiles identify miR-378 as a serum biomarker for early detection of gastric cancer. Cancer Lett. 2012;316(2):196-203. [DOI] [PubMed] [Google Scholar]
  • 84.Fei B, Wu H. MiR-378 inhibits progression of human gastric cancer MGC-803 cells by targeting MAPK1 in vitro. Oncol Res. 2012;20(12):557-564. [DOI] [PubMed] [Google Scholar]
  • 85.Hua Z, Lv Q, Ye W, et al. MiRNA-directed regulation of VEGF and other angiogenic factors under hypoxia. PloS One. 2006;1(1):e116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Li H, Dai S, Zhen T, et al. Clinical and biological significance of miR-378a-3p and miR-378a-5p in colorectal cancer. European Journal Of Cancer (Oxford, England : 1990). 2014;50(6): 1207-1221. doi: 10.1016/j.ejca.2013.12.010. [DOI] [PubMed] [Google Scholar]
  • 87.Peng J, Xie Z, Cheng L, et al. Paired design study by real-time PCR: miR-378* and miR-145 are potent early diagnostic biomarkers of human colorectal cancer. BMC Cancer. 2015;15:158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zanutto S, Pizzamiglio S, Ghilotti M, et al. Circulating miR-378 in plasma: A reliable, haemolysis-independent biomarker for colorectal cancer. Br J Cancer. 2014;110(4):1001-1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Tanaka H, Hazama S, Iida M, et al. miR-125b-1 and miR-378a are predictive biomarkers for the efficacy of vaccine treatment against colorectal cancer. Cancer Sci. 2017;108(11):2229-2238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Shindo Y, Hazama S, Nakamura Y, et al. miR-196b. miR. (378a), and miR-486 are predictive biomarkers for the efficacy of vaccine treatment in colorectal cancer. Oncol Lett. 2017;14(2):1355-11362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ye H, Li W, Wu K, et al. The SP1-induced long noncoding RNA, LINC00339, promotes tumorigenesis in colorectal cancer via the miR-378a-3p/MED19 axis. Onco Targets Ther. 2020;13:11711-111724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Yang J, Ma Q, Zhang M, Zhang W. LncRNA CYTOR drives L-OHP resistance and facilitates the epithelial-mesenchymal transition of colon carcinoma cells via modulating miR-378a-5p/SERPINE1. Cell Cycle (Georgetown, Tex). 2021;20(14):1415-1430. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 93.Zeng M, Zhu L, Li L, Kang C. miR-378 suppresses the proliferation, migration and invasion of colon cancer cells by inhibiting SDAD1. Cell Mol Biol Lett. 2017;22:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zheng Y, Liu Y, Lin Y, et al. MicroRNA-124 and microRNA-378 inhibit the proliferation and invasion of colorectal cancer by upregulating KiSS1. Transl Cancer Res. 2020;9(4):2838-2846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.An J, Liu J, Liu L, et al. A genetic variant in primary miR-378 is associated with risk and prognosis of hepatocellular carcinoma in a Chinese population. PloS One. 2014;9(4):e93707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Qian F, Wang J, Wang Y, et al. MiR-378a-3p as a putative biomarker for hepatocellular carcinoma diagnosis and prognosis: Computational screening with experimental validation. Clin Transl Med. 2021;11(2):e307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Zhu B, Chen J, Feng Y, et al. DNMT1-induced miR-378a-3p silencing promotes angiogenesis via the NF-κB signaling pathway by targeting TRAF1 in hepatocellular carcinoma. J Exp Clin Cancer Res: CR. 2021;40(1):352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Fu H, Zhang J, Pan T, Ai S, Tang L, Wang F. Mir-378a enhances the sensitivity of liver cancer to sorafenib by targeting VEGFR, PDGFRβ and c-Raf. Mol Med Rep. 2018;17(3):4581-4588. [DOI] [PubMed] [Google Scholar]
  • 99.Wang J, Li Y, Ma Q, Huang J. Mir-378 in combination with ultrasonic irradiation and SonoVue microbubbles transfection inhibits hepatoma cell growth. Mol Med Rep. 2020;21(6):2493-2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Ma J, Lin J, Qian J, et al. MiR-378 promotes the migration of liver cancer cells by down-regulating Fus expression. Cell Physiol Biochem: Int J Exp Cell Physiol, Biochem, Pharm. 2014;34(6):2266-2274. [DOI] [PubMed] [Google Scholar]
  • 101.Zou H, Yang L. miR-378a-5p improved the prognosis and suppressed the progression of hepatocellular carcinoma by targeting the VEGF pathway. Transl Cancer Res. 2020;9(3):1558-1566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lin Z, Xia S, Liang Y, et al. LXR Activation potentiates sorafenib sensitivity in HCC by activating microRNA-378a transcription. Theranostics. 2020;10(19):8834-8850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Li Y, Zhou T, Cheng X, Li D, Zhao M, Zheng W. microRNA-378a-3p regulates the progression of hepatocellular carcinoma by regulating PD-L1 and STAT3. Bioengineered. 2022;13(3):4730-4743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Ji Y, Yang S, Yan X, et al. CircCRIM1 promotes hepatocellular carcinoma proliferation and angiogenesis by sponging miR-378a-3p and regulating SKP2 expression. Front Cell Dev Biol. 2021;9:796686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Onishi M, Ichikawa T, Kurozumi K, Date I. Angiogenesis and invasion in glioma. Brain Tumor Pathol. 2011;28(1):13-24. [DOI] [PubMed] [Google Scholar]
  • 106.Xue S, Hu M, Li P, et al. Relationship between expression of PD-L1 and tumor angiogenesis, proliferation, and invasion in glioma. Oncotarget. 2017;8(30):49702-49712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Li W, Liu Y, Yang W, et al. MicroRNA-378 enhances radiation response in ectopic and orthotopic implantation models of glioblastoma. J Neuro-Oncol. 2018;136(1):63-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Shi H, Wang D, Sun X, Sheng L. MicroRNA-378 acts as a prognosis marker and inhibits cell migration, invasion and epithelial-mesenchymal transition in human glioma by targeting IRG1. Eur Rev Med Pharmacol Sci. 2018;22(12):3837-3846. [DOI] [PubMed] [Google Scholar]
  • 109.Li B, Wang Y, Li S, et al. Decreased expression of miR-378 correlates with tumor invasiveness and poor prognosis of patients with glioma. Int J Clin Exp Pathol. 2015;8(6):7016-7021. [PMC free article] [PubMed] [Google Scholar]
  • 110.Guo X, Zhang X, Chen P, Ma L, Shen Z. Mir-378a-3p inhibits cellular proliferation and migration in glioblastoma multiforme by targeting tetraspanin 17. Oncol Rep. 2019;42(5):1957-1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Deng Z, Du W, Fang L, et al. The intermediate filament vimentin mediates microRNA miR-378 function in cellular self-renewal by regulating the expression of the Sox2 transcription factor. J Biol Chem. 2013;288(1):319-331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Lee D, Deng Z, Wang C, Yang B. MicroRNA-378 promotes cell survival, tumor growth, and angiogenesis by targeting SuFu and Fus-1 expression. Proc Natl Acad Sci U S A. 2007;104(51):20350-20355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Avgeris M, Stravodimos K, Scorilas A. Loss of miR-378 in prostate cancer, a common regulator of KLK2 and KLK4, correlates with aggressive disease phenotype and predicts the short-term relapse of the patients. Biol Chem. 2014;395(9):1095-1104. [DOI] [PubMed] [Google Scholar]
  • 114.Cannistraci A, Hascoet P, Ali A, et al. MiR-378a inhibits glucose metabolism by suppressing GLUT1 in prostate cancer. Oncogene. 2022;41(10):1445-1455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Mao Y, Li W, Hua B, et al. Circular RNA_PDHX promotes the proliferation and invasion of prostate cancer by sponging MiR-378a-3p. Front Cell Dev Biol. 2020;8(602707). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Ikeda K, Horie-Inoue K, Ueno T, et al. miR-378a-3p modulates tamoxifen sensitivity in breast cancer MCF-7 cells through targeting GOLT1A. Sci Rep. 2015;5(13170). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Rong D, Dong Q, Qu H, et al. mA-induced LINC00958 promotes breast cancer tumorigenesis via the miR-378a-3p/YY1 axis. Cell Death Discov. 2021;7(1):27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Niu M, Shan M, Liu Y, et al. DCTPP1, an oncogene regulated by miR-378a-3p, promotes proliferation of breast cancer via DNA repair signaling pathway. Front Oncol. 2021;11(641931). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Zheng S, Li M, Miao K, Xu H. lncRNA GAS5-promoted apoptosis in triple-negative breast cancer by targeting miR-378a-5p/SUFU signaling. J Cell Biochem. 2020;121(3):2225-2235. [DOI] [PubMed] [Google Scholar]
  • 120.Gong W, Zhu C, Liu Y, et al. Elevated levels of both microRNA 378 (miR-378) and kallikrein-related peptidase 4 (KLK4) mRNA are associated with an unfavorable prognosis in triple-negative breast cancer. Am J Transl Res. 2021;13(3):1594-1606. [PMC free article] [PubMed] [Google Scholar]
  • 121.Scapoli L, Palmieri A, Lo Muzio L, et al. MicroRNA expression profiling of oral carcinoma identifies new markers of tumor progression. Int J Immunopathol Pharmacol. 2010;23(4):1229-1234. [DOI] [PubMed] [Google Scholar]
  • 122.Chen X, Yu J, Tian H, et al. Circle RNA hsa_circRNA_100290 serves as a ceRNA for miR-378a to regulate oral squamous cell carcinoma cells growth via glucose transporter-1 (GLUT1) and glycolysis. J Cell Physiol. 2019;234(11):19130-19140. [DOI] [PubMed] [Google Scholar]
  • 123.Li J, Sun S, Li J, et al. NCAPG, mediated by miR-378a-3p, regulates cell proliferation, cell cycle progression, and apoptosis of oral squamous cell carcinoma through the GSK-3β/β-catenin signaling. Neoplasma. 2021;68(6):1201-1211. [DOI] [PubMed] [Google Scholar]
  • 124.Cui Y, Xie M, Zhang Z. LINC00958 Involves in bladder cancer through sponging miR-378a-3p to elevate IGF1R. Cancer Biother Radiopharm. 2020;35(10):776-788. [DOI] [PubMed] [Google Scholar]
  • 125.Ding N, Sun X, Wang T, Huang L, Wen J, Zhou Y. Mir-378a-3p exerts tumor suppressive function on the tumorigenesis of esophageal squamous cell carcinoma by targeting Rab10. Int J Mol Med. 2018;42(1):381-391. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 126.Liu H, Zhang Q, Song Y, et al. Long non-coding RNA SLC2A1-AS1 induced by GLI3 promotes aerobic glycolysis and progression in esophageal squamous cell carcinoma by sponging miR-378a-3p to enhance Glut1 expression. J Exp Clin Cancer Res: CR. 2021;40(1):287. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 127.Wang X, Liu H, Zhang Q, et al. LINC00514 promotes lipogenesis and tumor progression in esophageal squamous cell carcinoma by sponging miR-378a-5p to enhance SPHK1 expression. Int J Oncol. 2021;59(5):86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Hummel R, Sie C, Watson D, et al. MicroRNA signatures in chemotherapy resistant esophageal cancer cell lines. World J Gastroenterol. 2014;20(40):14904-14912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Weng W, Leung W, Pang Y, Hsu H. Lauric acid can improve the sensitization of Cetuximab in KRAS/BRAF mutated colorectal cancer cells by retrievable microRNA-378 expression. Oncol Rep. 2016;35(1):107-116. [DOI] [PubMed] [Google Scholar]
  • 130.Weng W, Leung W, Pang Y, Kuo L, Hsu H. EPA Significantly improves anti-EGFR targeted therapy by regulating miR-378 expression in colorectal cancer. Oncol Lett. 2018;16(5):6188-6194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Wu Q, Xie Y, Deng Z, et al. Ergosterol peroxide isolated from Ganoderma lucidum abolishes microRNA miR-378-mediated tumor cells on chemoresistance. PloS One. 2012;7(8):e44579. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Technology in Cancer Research & Treatment are provided here courtesy of SAGE Publications

RESOURCES