- Research
- Open access
- Published:
Effect of conditioned medium from miRNA-34a transfected gastric cancer-associated fibroblast on peripheral blood mononuclear cells
BMC Immunology volumeĀ 26, ArticleĀ number:Ā 9 (2025)
Abstract
Background
Cancer-associated fibroblast (CAF) cells play an important role in gastric malignancy. MiRNA dysregulation has been detected in CAF cells, which is related to the tumor progression ability of these cells. Therefore, this study aimed to evaluate the function of miRNA34a in CAF cells in gastric carcinoma.
Method
We transiently transfected miRNA-34a mimic in CAF cells and examined the effect of the overexpressed miRNA on PD-L1 expression using real-time PCR. Next, we evaluated the role of transfected CAF-conditioned medium (CM) on the immune response and viability of gastric cancer cell lines.
Results
We have shown that miRNA-34a significantly reduced PD-L1 expression in CAF cells (pā<ā0.05). However, the conditioned medium of transfected cells had no significant effect on the immune response. We also found that CM of miRNA-34a transfected CAF cells significantly suppressed gastric cancer cell line viability relative to the control group (Pā<ā0.05).
Conclusion
We indicated that CM of miRNA-34a transfected CAF can reduce gastric cancer cell line proliferation. Additionally, miRNA-34a in these cells may improve immune response via PD-L1 reduction. Thus, miRNA-34a could be a potential therapeutic agent in gastric cancer treatment.
Clinical trial number
Not applicable.
Introduction
Gastric cancer is one of the most common malignancies and the leading cause of cancer-related death worldwide. In 2024, More than 26,000 new cases are diagnosed, and 10,880 patients died of this malignancy in the United States [1, 2]. The Survival rates for patients with advanced-stage gastric cancer remain low despite chemotherapy, surgery, hormone therapy and other treatments [3,4,5].
The tumor microenvironment (TME) has an important role in cancer progression. TME consists of the extracellular matrix (ECM), blood vasculature, inflammatory cells, immune cells and cancer-associated fibroblast cells [6, 7]. Cancer-associated fibroblast (CAF) cells are the most important and prominent cells in the tumor stroma. These cells can increase tumor invasion, metastasis, and angiogenesis and have an immunosuppressive role in malignancies via the secretion of various mediators and direct interaction with tumor cells and immune cells [8,9,10].
98% of the genome includes noncoding RNA, such as microRNA (miRNA). MiRNA āsmall noncoding RNAsā contain 19ā25 nucleotides that can interact with the 3ā untranslated region (3āUTR) of their target gene mRNA and inhibit the protein expression [11, 12]. MiRNAs play a pivotal role in transcriptional and post-transcriptional levels and are related to many biological activities and diseases, such as cell apoptosis, differentiation, metabolism, proliferation, blood pressure, cardiac function, autoimmune disease, viral infection and different types of malignancies. These molecules exhibit an oncogenic or tumor suppressor role in cancers, depending on their target genes [13,14,15,16].
MiRNA dysregulation in CAFs, contributes to the tumor progression ability of these cells. MiRNA molecules can influence the reprogramming of normal fibroblasts into CAFs and their function. Also, secreted miRNA from CAF may influence several aspects of a cancer cellās behavior [17,18,19,20,21].
MiRNA-34a is a tumor suppressor miRNA that is down-regulated in numerous types of malignancies, such as gastric cancer, colorectal cancer, and breast cancer. This miRNA involves post- transcriptional regulation of numerous genes such as TGF-βR1, β-catenin, PD-L1, and NOTCH1 that have an effect on tumor cell proliferation, invasion, epithelial-mesenchymal transition (EMT) and immune response [22].
Studies have shown that miRNA can be used as a therapeutic target in malignancies [23]. CAF cells and their secreted exosomes in oral squamous cell carcinoma (OSCC) contain a low level of miRNA-34a, and transfection of this molecule into CAF cells can reduce the tumor genesis of OSCC cells. Therefore, according to previous studies, overexpression of miRNA-34a into CAF can be considered as a possible therapeutic strategy [24].
In this present study, miRNA-34a mimic was transfected into gastric CAF (GC-CAF) cells. Next, we evaluated the effect of miRNA-34a mimic overexpression in these cells on PD-L1 expression as an immune checkpoint molecule. Furthermore, the conditioned media of GC-CAF cells were collected after miRNA transfection. Then, we measured the effect of miRNA-34a transfected GC-CAF conditioned media on the immune response and viability of gastric cancer cell lines. The result of this study may provide new insight into more efficient strategies in cancer therapy.
Material and method
Sample collection
Tumor samples were obtained under sterile conditions from untreated patients with gastric cancer during an endoscopic examination at the Tuba Clinic (Mazandaran, Iran) (the informed consent was obtained from all of the participants). Sterile tubes containing Dulbeccoās Modified Eagle Medium F12 (DMEM F12) supplemented with 10% penicillin/streptomycin (Biowest) and 10% amphotericin B (Capricorn, AAS-b, Republic of Germany) were used to transfer the samples. Samples were processed within a maximum time of 24Ā h after resection.
CAF isolation
Gastric cancer tissue samples were washed in PBS containing 10% penicillin/streptomycin and 10% amphotericin B for 30Ā min to remove yeast and bacterial contamination. Subsequently, the samples were cut into small pieces. Tissue fragments were digested with collagenase IV (1.5Ā mg/ml) (Gibco Cat, No: 17104-D19) at 37\(\:ā\) for 35Ā min. Next, the suspensions were plated in DMEM F12 with 20% fetal bovine serum (FBS) (Biosera), 1% penicillin/streptomycin, and 1% amphotericin B. After 24Ā h, the medium was replaced with a fresh medium. After that, the medium was changed every 3 days. Cells typically appeared within one to two weeks after tissue culture.
CAFs are more sensitive than epithelial cells to trypsin; therefore, differential trypsinization was used to separate and purify cancer-associated fibroblast cells from cancer epithelial cells [25]. The cells were characterized by assessing of αSMA (alpha-smooth muscle actin) and EPCAM (Epithelial cell adhesion molecule) expression using real-time PCR. All cells used in this research were from 3 to 7 passages.
Cell lines culture
AGS and Kato3 (gastric cancer cell lines) were cultured in RPMI supplemented with 10% FBS and 1% penicillin/streptomycin under optimal conditions of 37\(\:ā\) and 5% CO2. Additionally, HGF, a human gingival normal fibroblast cell line, was cultured in DMEM F12 with 10%FBS and 1% penicillin/streptomycin at 37\(\:ā\) and 5% CO2.
RNA extraction and reverse transcription (cDNA synthesis)
Total RNA was extracted from cells using Trizol reagent (one-step RNA reagent, Bio Basic, Cat. No: BS410A) according to the manufacturerās instructions. The total RNA concentration was measured using a PicoDrop spectrophotometer.
The extracted RNA was treated with RNase free -DNase I (Thermo Scientific, #EN0521) according to the manufacturerās protocol to remove DNA contamination. CDNA synthesis was performed using a cDNA synthesis kit (Yektatajhiz Azma (YTA), Cat No: YT4500). MiRNA-34a stem-loop primer and U6-specific primer were used for the reverse transcription of miRNA-34a and U6 RNA, respectively. CDNA synthesis from other gene transcripts was performed using Oligo dT and random hexamer primers.
Quantitative real time (qRT) -PCR-
Quantification of miRNA-34a, αSMA, EPCAM, and PD-L1 transcript levels was performed by qRT-PCR analysis using SYBR Green qPCR mastermix 2x (Yektatajhiz Azma (YTA), Cat No: YT2551).
The U6 reference gene was used to normalize miRNA-34a expression, while the GAPDH reference gene was employed to normalize the expression of the other genes. Primers sequences are shown in TableĀ 1.
Transfection of miRNA-34a mimic into CAF cells
CAF cells were seeded in the six-well plates at a density of 2āĆā105 cells per well in DMEM F12ā+ā20% FBSā+ā1% penicillin/streptomycin medium. After 24Ā h, the medium was replaced with an antibiotic-free medium (DMEM F12ā+ā10% FBS). The cells were transfected with the miRNA-34a mimic or miR negative control (NC) at a 20 nM concentration, using Lipofectamin2000 (Invitrogen) according to the manufacturerās instructions. RT qPCR was performed to verify the transfection efficiency 48Ā h post-transfection.
Preparation CAF conditioned medium (CM)
CAF cells were seeded in the six-well plate at a density of 2āĆā105 cells per well and were transfected with miRNA34a mimic or miR negative control (NC) at 20nM concentration. Twenty-four hours after transfection, the culture medium was replaced with a new medium containing DMEM F12 without FBS in each well. After 48Ā h, the supernatant from each group was harvested and centrifuged at 3000Ā rpm for 10Ā min to remove cell debris. The conditioned medium was stored at -80 \(\:ā\) for future analysis.
Measurement of protein concentration in CAFās CM
The total protein concentration of CAFās CM in the transfected and NC groups was determined by the Bradford test using the Better Bradford Protein Assay Kit (Bio Basic INC, Cat. No: SK3041) according to the manufacturerās instructions.
Measurement of cytokine concentration using enzyme-linked immunosorbent assay (ELISA)
Peripheral blood mononuclear cells (PBMCs), from healthy donors (n:3) were activated with 1 µg/ml PHA and treated with miRNA-34a-transfected CAF CM or NC group CAF CM at the optimal protein concentration of 2.265 µg/ml for 72Ā h. Subsequently, the supernatant of the cells was collected, and concentrations of IL-4 and IFN-\(\:\gamma\:\) were determined using an ELISA kit (Parsgene) according to the manufacturerās instructions.
Cytotoxic T cells population measurements using flow cytometry method
The percentage of the CD8ā+āT cell population in PBMC cells from healthy donors was evaluated by flow cytometry after treatment with CAF CM in different groups. For this purpose, 72Ā h post-treatment, the PBMC cells were collected by centrifugation at 300Ā g for five minutes. Subsequently, the cells were stained with dual-color antibodies: anti-human CD3-FITC and anti-human CD4 RPE (Dakocytomation, cat. no FR881) in 100µL of phosphate buffered saline (PBS) supplemented with 1% Bovine Serum Albumin (BSA) according to the manufacturerās instructions. Next, the CD8 and CD3 double-positive cells were determined using flow cytometry. The results were analyzed withe FlowJo software 7.6.1 (Tree Star, Inc., San Carlos, CA, USA).
Cells viability assay
The viability of gastric cancer cell lines (AGS and Kato3) was assessed using the methyl thiazolyl tetrazolium test (MTT). Briefly, cancer cells were seeded in complete media (DMEMF12ā+ā10% FBSā+ā1% penicillin/streptomycin) at 2.5āĆā103 cells/well in 96-well plates. Twenty-four hours later, the cells were treated with miRNA-34a-transfected CAF CM and NC CAF CM at the optimal protein concentration of 2.265 µg/ml for 48Ā h. Next, MTT solution at a final concentration of 0.5Ā mg/ml was added to each well, and incubation was performed for four hours at 37\(\:ā\). At the end of the incubation time, the medium was removed, and 200 µl of DMSO was added to dissolve the formazan crystal. Next, the plate was shaken for ten minutes. Finally, the absorbance of each well was measured using a microplate reader at 570Ā nm with a reference of 630Ā nm.
Additionally, we evaluated PBMC viability 72Ā h after treatment with CAF cell CM using the MTT test according to the above instructions.
Statistical analysis
An unpaired T-test was used to determine the statistical differences between the two groups. The unpaired ANOVA test was used to determine the statistical differences between the three groups. Normal data were expressed as meanā±āSD. P-valuesā<ā0.05 were considered significant. Statistical analysis of data and plotting of graphs were performed using GraphPad Prism software version 6.
Result
Characterization of gastric cancer-associated fibroblast
We isolated primary gastric cancer-associated fibroblast cells from the gastric cancer tissue of a patient who had undergone an endoscopic operation in a sterile conditions. The CAF cells exhibited spindle-like morphology (Fig.Ā 1a). The expression of αSMA (fibroblast activation marker) was significantly higher in CAF cells than in the human normal gingival fibroblast (HGF) cell line (Pā<ā0.05). Additionally, neither of the cells (CAF and HGF cell line) expressed the EPCAM marker as an epithelial indicator, suggesting the successful isolation of CAF cells (Fig.Ā 1b).
Overexpression of miRNA-34a in CAF cells decreased PD-L1 expression
We transiently transfected the miRNA-34a mimic into CAF cells. Forty eight hours post- transfections, we evaluated the transfection efficacy. The qRT-PCR results showed that miRNA-34a was overexpressed in these cells in comparison with the NC groups (P: 0.0027) (Fig.Ā 2a). Next, we assessed the effect of miRNA-34a overexpression on its target gene, PD-L1, in CAF cells. MiRNA-34a could significantly reduced PD-L1 expression in CAF cells compared to the NC group (P: 0.0001) (Fig.Ā 2b).
miRNA-34a-transfected CAF CM did not affect the immune response
We measured the effect of CM from miRNA-34a-transfected CAF cell on PBMC proliferation, the percentage of CD8-positive T cells, and cytokine (IFN-γ and IL-4) secretion ability 72 h after treatment.
The PBMCs were activated with PHA (1 µg/ml) and treated with miRNA-34a-transfected CAF CM (test group) or miR negative control-transfected CAF CM (NC CAF CM) (control 2) for 72 h and results were compared with data from the group without CM treatment (Control 1).
Our observation showed that cytokine secretion (IL-4 and IFN-γ) and PBMC viability slightly higher in the control 2 group relative to the others (test and control 1), but the differences were not statistically significant(Pā>ā0.05) (Fig.Ā 3a, b,c). In addition, miRNA-34a-transfected CAF CM did not influence the percentage of CD8-positive T cell population (Test) (24.5ā±ā3.928) compared with the two other groups, the non-treated group (control 1) (23.65ā±ā5.62) and the treated with NC CAF CM (control 2) (25.63ā±ā2.281) (P:0.839) (Fig.Ā 4a, b,c, d).
Effect of CM from miRNA-34a transfected CAF on PBMCs cytokine secretion and viability. a, b: There wasnāt significant differences in IFN gamma and IL-4 concentrations detected in PBMC supernatant after treatment with miRNA-34a transfected CAF and NC CAF CM (a: P:0.0183, b: p:0.339) c: There werenāt differences in PBMC viability among different groups (P:0.087). Control 1: PBMC+ PHA, Control 2: PBMC+PHA+NC CAF CM, Test: PBMC+PHA+miRNA-34a transfected CAF CM
Effect of miRNA-34a transfected CAF conditioned medium on percentage of cytotoxic T cell population. a, b, c, d: Forty-eight hours after treatment PBMCs with CAF conditioned medium we evaluated CD8 positive T cell populations in different groups. CM from miRNA-34a transfected CAF didnāt influence on percentage of CD8 positive T cell population (Test) (24.5 ± 3.928) compared with two other groups, control 1 (23.65 ± 5.62) and control 2 (25.63 ± 2.281) (P:0.839). Control 1: PBMC+ PHA, Control 2: PBMC+NC CAF CM, Test: PBMC+miRNA-34a transfected CAF CM
miRNA-34a-transfected CAF CM reduced gastric cancer cell lines viability
To determine the effect of CM from miRNA-34a-transfected CAF on the viability of gastric cancer cells, AGS and KATO3 cell lines were treated with CAF CM at optimal protein concentrations. Cell viability was measured using the MTT assay. The results revealed that NC CAF CM could significantly increase AGS cell line viability relative to the group without CM treatment (Fig.Ā 5a, P:0.03). It also, improved KATO3 cell line viability, but the effect was not significant (Fig.Ā 5b, P: 0.8). MiRNA-34a-transfected CAF CM significantly suppressed viability in AGS cell line compared to groups treated with NC CAF CM (Fig.Ā 5a, Pā<ā0.05, P:0.001) and the control group (Fig.Ā 5a, P: 0.005). It also significantly reduced the viability of KATO3 cell lines relative to the group treated with NC CAF CM (Fig.Ā 5b, P: 0.019).
a, b: NC CAF CM could significantly increase AGS cell lines viability relative to control group without CM treatment (P:0.03). Also NC CAF CM increased Kato3 cell lines viability but itās not a significant (P: 0.8). MiRNA-34a transfected CAF CM could significantly suppress viability in AGS cell lines compared with groups treated with NC CM (P:0.001) and control group (P: 0.005). Also it could significantly reduce Kato3 cell lines viability compared with group treated with NC CM (P: 0.019)
Discussion
Our study is divided into two parts. In the first part, we measured the effect of miRNA-34a transfection in CAF cells on immune responses by evaluating of PD-L1 expression in these cells and the effect of conditioned media from miRNA-34a-transfected CAF on the CD8 -positive T cell population, as well as the secretion of IFN-γ and IL-4 cytokines in vitro.
Our results showed that miRNA-34a transfection in CAF cells decreased PD-L1 expression in these cells (Pā<ā0.05) (Fig.Ā 2b).
Tumor cells use different mechanisms to suppress the immune response by inducing T cell exhaustion through the upregulation of immune checkpoint molecules, including, CTLA-4, LAG-3, PD-L1, ITIM and TIGIT [26].
PD-L1 is an immune checkpoint ligand that is often expressed in tumor cells. The binding of PD-L1 to PD-1 on T cells, activates inhibitory signals that can suppress T cell activity. Prevention of PD-1 and PD-L1 binding, leads to improved cytotoxic T cell function against tumor cells in several malignancies [27].
CD8+ T cells play an important role in antitumor immunity. These cells recognize tumor cells via HLA class I, secreting cytokines such as TNF- α and IFN-γ, and releasing perforin and granulysin to eradicate tumor cells [28, 29].
MiRNA-34a targets PD-L1 mRNA, resulting in a reduction of PD-L1 [30, 31]. Additionally, the Epstein-Barr virus can increase PD-L1 expression by downregulating miRNA-34a in B-cell lymphoma [32].
PD-L1 expression in CAF cells suppresses the immune response and contributes to the poor prognosis and outcomes in several malignancies, such as esophageal cancer and non-small cell lung carcinoma [33, 34].
IL-1α/β from tumor cells in melanoma, induced PD-L1 and PD-L2 upregulation in CAF cells, contributing to the suppression of the immune response [35].
PD-L1 and PD-L2 molecules overexpression in fibroblast interact with the PD-1 receptors directly, and induce T cells exhaustion that leads CD8+ T cells function abrogating [34,35,36]. Therefore, miRNA-34a in CAF cells may enhance the immune response by reducing PD-L1 expression.
CAF cells can influence the tumor microenvironment and immune response through the secretion of various factors. Therefore, we also evaluated the effect of conditioned medium from miRNA-34a- transfected CAF cells on the immune response. After treating of PBMCs with CAF CM, the inhibitory effect of CAF CM on the immune response was not observed. Additionally, CM from miRNA-34a-transfected CAFs exhibited a slight inhibitory effect on PBMCs proliferation, cytokine secretion (IFN-γ, IL-4), and the percentage of CD8-positive T cells population. Surprisingly, the immune response was slightly more stimulated in a group treated with NC CAF CM than in, others, but it wasnāt significant (Figs.Ā 3a, b and c and 4a and b).
Previous investigations have shown that CAF cells play an important role in T cell polarization, proliferation, cytokine secretion, and possess an immunosuppressive effect through, both direct and indirect pathways [36]. Gorchs and colleagues reported that CAF cells from lung carcinoma maintain their immunosuppressive effect even after high-dose radiation [37]. Conversely, Nazareth and coworkers identified heterogeneous CAF populations in non-small cell lung carcinoma with a different effects on the immune response; one cell subpopulation can suppress the immune response via PD-L2 and PD-L1 expression and TGF-β secretion, while another cell subpopulation can enhance the immune response through direct pathways. However, the supernatant from these cells did not significantly influence the immune response [38].
MRX-34, a liposomal form of miRNA-34a, is the first miRNA to reach a Phase I clinical trial in cancer therapy. Results have shown that MRX-34, in addition to decreasing PD-L1 expression, can also increase CD8+ T cell infiltration and reduce regulatory T cell and macrophage infiltration in the tumor stroma. Moreover, suppressing the miRNA-34a expression in HBV+ - hepatocellular carcinoma accelerates the production of CCL22, which enhances regulatory T cell infiltration in the tumor stroma [39, 40].
Conversely, a study showed that the overexpression of miRNA-34a in T cells can inhibit proliferation, activation, and effector function by modulating the NF- ĪŗB signaling pathway in these cells [41]. However, in the present study, significant differences were not detected in experiments related to the treatment of PBMCs with CM from miRNA-34a-transfected CAF cells compared to the control group.
In the second part, we evaluated the effect of CM from miRNA-34a transfected CAF cells on the proliferation of gastric cancer (GC) cell lines.
CAF cells contribute to the tumor progression by enhancing cancer cell invasion, metastasis, and angiogenesis through the secretion of various growth factors and ECM (extracellular matrix) degrading proteases [9, 42,43,44,45].
In this part, we have revealed that the treatment of gastric cancer cell lines (AGS, Kato3) with CM from miRNA-34a-transfected CAF significantly suppressed the proliferation rate of these cell lines (Fig.Ā 5a and b). In previous reports, the transfection of miRNA-34a in gastric CAF and oral squamous cell carcinoma CAF could increase miRNA-34 in the secreted-exosomes of these cells. Subsequently, miRNA-34a transfected CAF cells can inhibit cancer cell growth and invasion via the transfer of miRNA34a to cancer cells through exosome secretion in-vitro and in-vivo [24, 45]. In contrast to the aforementioned studies, the present study used the supernatant of CAF cells which it contains exosomes and other additional molecules. Therefore, in our research, the effect of CAF CM may not be merely due to the secreted exosomes. In other words, the efficient role of other unknown molecules is inevitable for achieving these results.
The presence of CAF cells in the TME is associated with the poor prognosis of malignancies, Depletion of these cells in pancreatic cancers correlates with immune suppression due to an increased regulatory T cell population in the tumor stroma and reduces patient survival. Additionally, in the early stages of malignancy, normal fibroblast cells can inhibit tumor progression via paracrine signaling through soluble factors, cellā to - cell contact, and ECM-integrity [46, 47]. Therefore, complete removal of these cells is not a good option in cancer treatment. Likely, the best approach is to find a way to reprogram these cells. In this study, we showed that miRNA-34a in CAF cells can reduce tumor cell viability, and probably improve the immune response in malignancies by suppressing PD-L1 expression in CAF cells. Our results indicate that miRNA-34a may have the potential to serve as a novel therapeutic strategy in cancer treatment.
Data availability
All data generated or analyzed during this study are available from the corresponding author.
Abbreviations
- CAF:
-
Cancer associated fibroblast
- miRNA:
-
microRNA
- PD-L1:
-
Programmed cell death ligand 1
- CM:
-
Conditioned medium
- TME:
-
Tumor microenvironment
- ECM:
-
Extra-cellular matrix
- UTR:
-
Untranslated region
- EMT:
-
Epithelial-mesenchymal transition
- OSCC:
-
Oral squamous cell carcinoma
- GC CAF:
-
Gastric CAF
- DMEM F12:
-
Dulbeccoās Modified Eagle Medium F12
- FBS:
-
Fetal bovine serum
- αSMA:
-
αsmooth muscle actin
- EPCAM:
-
Epithelial cell adhesion molecule
- HGF:
-
Human gingival fibroblast
- PBMC:
-
Peripheral blood mononuclear cell
- NC:
-
Negative control
- PBS:
-
Phosphate buffer saline
- BSA:
-
Bovine serum albumin
- MTT:
-
Methyl thiazolyl tetrazolium
References
Lin JL, Lin JX, Lin GT, Huang CM, Zheng CH, Xie JW, et al. Global incidence and mortality trends of gastric cancer and predicted mortality of gastric cancer by 2035. BMC Public Health. 2024;24(1):1763.
Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. Cancer J Clin. 2024;74(1):12ā49.
Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Cancer J Clin. 2018;68(6):394ā424.
Rawla P, Barsouk A. Epidemiology of gastric cancer: global trends, risk factors and prevention. Przeglad Gastroenterologiczny. 2019;14(1):26ā38.
Sonkin D, Thomas A, Teicher BA. Cancer treatments: past, present, and future. Cancer Genet. 2024;286ā287:18ā24.
Arneth B, Kaunas. Lithuania). 2019;56(1).
Bhowmick NA, Neilson EG, Moses HL. Stromal fibroblasts in cancer initiation and progression. Nature. 2004;432(7015):332ā7.
Ishii G, Ochiai A, Neri S. Phenotypic and functional heterogeneity of cancer-associated fibroblast within the tumor microenvironment. Adv Drug Deliv Rev. 2016;99(Pt B):186ā96.
LeBleu VS, Kalluri R. A peek into cancer-associated fibroblasts: origins, functions and translational impact. Dis Models Mech. 2018;11(4).
Tao L, Huang G, Song H, Chen Y, Chen L. Cancer associated fibroblasts: an essential role in the tumor microenvironment. Oncol Lett. 2017;14(3):2611ā20.
OāBrien J, Hayder H, Zayed Y, Peng C. Overview of MicroRNA Biogenesis, mechanisms of actions, and circulation. Front Endocrinol. 2018;9:402.
Si W, Shen J, Zheng H, Fan W. The role and mechanisms of action of microRNAs in cancer drug resistance. Clin Epigenetics. 2019;11(1):25.
Gou Z, Li J, Liu J, Yang N. The hidden messengers: cancer associated fibroblasts-derived exosomal miRNAs as key regulators of cancer malignancy. Front cell Dev Biology. 2024;12:1378302.
Peng Y, Croce CM. The role of MicroRNAs in human cancer. Signal Transduct Target Therapy. 2016;1:15004.
Seyhan AA. Trials and tribulations of MicroRNA therapeutics. Int J Mol Sci. 2024;25(3).
Shaheen N, Shaheen A, Diab RA, Desouki MT. MicroRNAs (miRNAs) role in hypertension: pathogenesis and promising therapeutics. Annals of medicine and surgery (2012). 2024;86(1):319āā28.
Ali S, Suresh R, Banerjee S, Bao B, Xu Z, Wilson J, et al. Contribution of microRNAs in understanding the pancreatic tumor microenvironment involving cancer associated stellate and fibroblast cells. Am J cancer Res. 2015;5(3):1251ā64.
Eichelmann AK, Matuszcak C, Hummel R, Haier J. Role of miRNAs in cell signaling of cancer associated fibroblasts. Int J Biochem Cell Biol. 2018;101:94ā102.
Pan Z, Tian Y, Niu G, Cao C. Role of microRNAs in remodeling the tumor microenvironment (review). Int J Oncol. 2020;56(2):407ā16.
Schoepp M, Strƶse AJ, Haier J. Dysregulation of miRNA expression in Cancer Associated fibroblasts (CAFs) and its consequences on the Tumor Microenvironment. Cancers. 2017;9(6).
Wang R, Sun Y, Yu W, Yan Y, Qiao M, Jiang R, et al. Downregulation of miRNA-214 in cancer-associated fibroblasts contributes to migration and invasion of gastric cancer cells through targeting FGF9 and inducing EMT. J Experimental Clin cancer Research: CR. 2019;38(1):20.
Zhang L, Liao Y, Tang L. MicroRNA-34 family: a potential tumor suppressor and therapeutic candidate in cancer. J Experimental Clin cancer Research: CR. 2019;38(1):53.
Rupaimoole R, Slack FJ. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases. Nat Rev Drug Discovery. 2017;16(3):203ā22.
Li YY, Tao YW, Gao S, Li P, Zheng JM, Zhang SE, et al. Cancer-associated fibroblasts contribute to oral cancer cells proliferation and metastasis via exosome-mediated paracrine miR-34a-5p. EBioMedicine. 2018;36:209ā20.
Wang X, Zhang W, Sun X, Lin Y, Chen W. Cancer-associated fibroblasts induce epithelial-mesenchymal transition through secreted cytokines in endometrial cancer cells. Oncol Lett. 2018;15(4):5694ā702.
Chen Y, Yu D, Qian H, Shi Y, Tao Z. CD8(+) T cell-based cancer immunotherapy. J Translational Med. 2024;22(1):394.
Parvez A, Choudhary F, Mudgal P, Khan R, Qureshi KA, Farooqi H, et al. PD-1 and PD-L1: architects of immune symphony and immunotherapy breakthroughs in cancer treatment. Front Immunol. 2023;14:1296341.
Li J, Han T, Wang X, Wang Y, Yang R, Yang Q. Development of a CD8ā+āT cell associated signature for predicting the prognosis and immunological characteristics of gastric cancer by integrating single-cell and bulk RNA-sequencing. Sci Rep. 2024;14(1):4524.
Preeti Rout FC. Zahid Iqbal, Prasanna Tadi. Histology,cytotoxic T Cells. 2023.
Wang X, Li J, Dong K, Lin F, Long M, Ouyang Y, et al. Tumor suppressor miR-34a targets PD-L1 and functions as a potential immunotherapeutic target in acute myeloid leukemia. Cellular signalling. 2015;27(3):443āā52.
Zuo Y, Zheng W, Liu J, Tang Q, Wang SS, Yang XS. MiR-34a-5p/PD-L1 axis regulates cisplatin chemoresistance of ovarian cancer cells. Neoplasma. 2020;67(1):93ā101.
Anastasiadou E, Stroopinsky D, Alimperti S, Jiao AL, Pyzer AR, Cippitelli C, et al. Epstein-Barr virus-encoded EBNA2 alters immune checkpoint PD-L1 expression by downregulating miR-34a in B-cell lymphomas. Leukemia. 2019;33(1):132āā47.
Kawasaki K, Noma K, Kato T, Ohara T, Tanabe S, Takeda Y, et al. PD-L1-expressing cancer-associated fibroblasts induce tumor immunosuppression and contribute to poor clinical outcome in esophageal cancer. Cancer immunology, immunotherapy: CII. 2023;72(11):3787āā802.
Teramoto K, Igarashi T, Kataoka Y, Ishida M, Hanaoka J, Sumimoto H, et al. Clinical significance of PD-L1-positive cancer-associated fibroblasts in pN0M0 non-small cell lung cancer. Lung cancer (Amsterdam, Netherlands). 2019;137:56ā63.
Khalili JS, Liu S, RodrĆguez-Cruz TG, Whittington M, Wardell S, Liu C, et al.Oncogenic BRAF(V600E) promotes stromal cell-mediated immunosuppression via induction of interleukin-1 in melanoma. Clinical cancer research: an official journal of the American Association for Cancer Research. 2012;18(19):5329-40.
An Y, Liu F, Chen Y, Yang Q. Crosstalk between cancer-associated fibroblasts and immune cells in cancer. Journal of cellular and molecular medicine. 2020;24(1):13ā24.
Gorchs L, Hellevik T, Bruun JA, Camilio KA, Al-Saad S, Stuge TB, et al. Cancer-associated fibroblasts from lung tumors maintain their immunosuppressive abilities after high-dose irradiation. Frontiers in oncology. 2015;5:87.
Nazareth MR, Broderick L, Simpson-Abelson MR, Kelleher RJ, Jr., Yokota SJ, Bankert RB. Characterization of human lung tumor-associated fibroblasts and their ability to modulate the activation of tumor-associated T cells. Journal of immunology (Baltimore,Md: 1950). 2007;178(9):5552-62.
Cortez MA, Anfossi S, Ramapriyan R, Menon H, Atalar SC, Aliru M, et al. Role of miRNAs in immune responses and immunotherapy in cancer. Genes, chromosomes & cancer.2019;58(4):244āā53.
Yang P, Li QJ, Feng Y, Zhang Y, Markowitz GJ, Ning S, et al. TGF-β-miR-34a-CCL22 signaling-induced Treg cell recruitment promotes venous metastases of HBV-positive hepatocellular carcinoma. Cancer cell. 2012;22(3):291ā303.
Hart M, Walch-Rückheim B, Friedmann KS, Rheinheimer S, Tänzer T, Glombitza B,et al. miR-34a: a new player in the regulation of T cell function by modulation of NF-κB signaling. Cell death & disease. 2019;10(2):46.
De Wever O, Demetter P, Mareel M, Bracke M. Stromal myofibroblasts are drivers of invasive cancer growth. International journal of cancer. 2008;123(10):2229-38.
Stuelten CH, DaCosta Byfield S, Arany PR, Karpova TS, Stetler-Stevenson WG, Roberts AB. Breast cancer cells induce stromal fibroblasts to express MMP-9 via secretion of TNF-alpha and TGF-beta. Journal of cell science. 2005;118(Pt 10):2143-53.
Unterleuthner D, Neuhold P, Schwarz K, Janker L, Neuditschko B, Nivarthi H, et al. Cancer-associated fibroblast-derived WNT2 increases tumor angiogenesis in colon cancer. Angiogenesis. 2020;23(2):159āā77.
Shi L, Wang Z, Geng X, Zhang Y, Xue Z. Exosomal miRNA-34 from cancer-associated fibroblasts inhibits growth and invasion of gastric cancer cells in vitro and in vivo.Aging. 2020;12(9):8549-64.
Alkasalias T, Moyano-Galceran L, Arsenian-Henriksson M, Lehti K. Fibroblasts in the Tumor Microenvironment: Shield or Spear? International journal of molecular sciences.2018;19(5).
Ćzdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, et al.Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer cell. 2014;25(6):719āā34.
Acknowledgements
This research was supported by the grant from mazandran-university of medical science, sari, Iran.
Funding
Not applicable.
Author information
Authors and Affiliations
Contributions
M E, performed whole research and wrote the manuscript N J, performed part of experiment and editing manuscript F A, data analysis S M V T, sample resection S A, Corresponding author.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Our study adhered to the declaration of Helsinki. the study protocol was approved by the Ethics Committee at mazandaran University of Medical Sciences (Ethics Code: IR.RUMS.REC.1400.031) and we were obtained informed consent to participate from all of the participants.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisherās note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the articleās Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the articleās 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-nc-nd/4.0/.
About this article
Cite this article
Esmaili, M., Jafari, N., Ahmadzadeh, F. et al. Effect of conditioned medium from miRNA-34a transfected gastric cancer-associated fibroblast on peripheral blood mononuclear cells. BMC Immunol 26, 9 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12865-025-00688-6
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12865-025-00688-6