Skip to main content

Exosomes-mediated transfer of LINC00691 regulates the formation of CAFs and promotes the progression of gastric cancer

Abstract

Objective

Gastric cancer (GC) is one of the malignant tumors with the highest mortality worldwide. Our previous studies have revealed that LINC00691 is up-regulated in serum of GC patients as a novel potential biomarker for GC diagnosis and prognosis. However, the roles of serum exosomal LINC00691 in GC has not been clarified. This study aimed to find the expression pattern of serum exosomal LINC00691 in GC patients and the correlation between the level of serum exosomal LINC00691 and the pathology of gastric cancer patients.

Methods

We collected the serum of 94 GC patients before surgery and extracted exosomes to detect the expression level of exosomal LINC00691, with 21 healthy volunteers and 17 patients with benign gastric diseases as controls. Surgical GC tissues and paired healthy tissues were collected to culture primary cancer-associated fibroblasts (CAFs) and normal fibroblasts (NFs). We then treated NFs with LINC00691-rich GC cell culture supernatant or exosomes and detected the activation markers and biological functions of the fibroblasts.

Results

The results of real-time qPCR indicated that the serum exosomal LINC00691 of GC patients was significantly higher than that of healthy subjects and patients with benign gastric diseases, and was associated with the clinicopathology of GC patients. More interestingly, when the NFs were treated with GC exosomes, the level of LINC00691 was significantly increased, the cell proliferation and migration were noticeably enhanced, and the ability to accelerate GC cell proliferation and invasion was promoted, which means that the induced fibroblasts gained the properties of CAFs. In addition, we found that knockdown of LINC00691 and the use of the JAK2/STAT3 signaling pathway inhibitor ruxolitinib effectively deprived exosome-containing GC cell supernatants of the effects on NFs.

Conclusion

Our study suggested that exosomal LINC00691 promoted NFs to gained the properties of CAFs depending on JAK2/STAT3 signaling pathway as a potential diagnostic biomarker for GC.

Peer Review reports

GC is the fifth most common malignant tumor and the third leading cause of cancer-related mortality worldwide, causing nearly 800,000 deaths every year [1, 2]. Early diagnosis of GC is a crucial prerequisite for effective clinical intervention. Pathological tissue biopsy is the gold method for definite diagnosis of GC although it is not appropriate to take it as a screening method in population because of invasive damage. Peripheral blood biomarkers, especially CEA, CA72-4, and CA19-9, are classical clinical screening means for GC [3]. However, the area under the curve (AUC) of the receiver operating characteristic curve (ROC) is respectively 0.653, 0.685, and 0.639, which is not satisfactory clinically [4]. Therefore, it is badly in need to search for new ideal biomarkers for GC diagnosis and prognosis [5, 6].

Systematic analysis studies have shown that the non-coding genome accounts for approximately 98% of all sequences, the aberrant expression of which is the main cause raising the transformation of tumor phenotype [7]. Long non-coding RNAs (lncRNAs) mainly refer to RNAs with more than 200 nucleotides and do not code proteins [8]. However, in recent years, a series of lncRNAs have been proved to produce small peptides [9]. In our previous study, we found that LINC00691 level was associated with tumor size, lymph node metastasis, invasion depth, and prognosis of patients. LINC00691 was overexpressed in GC and the ROC curve was better than traditional classical indicators [10]. In this study, we found the AUC of serum exosomal LINC00691 even larger than serum LINC00691.

Exosomes are vesicles secreted by cells that can carry biologically active substances [11, 12]. Studies have shown that exosomes from tumor cells regulate cell reprogramming, thereby promoting the transformation of NFs to CAFs. Moreover, this effect may rely on the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway [13]. We previously reported that LINC00691 promoted GC cell proliferation and invasion through the JAK2/STAT3 signaling pathway [10]. The JAK/STAT signaling pathway has been reported as a common pathway in the regulation of many cellular biological activities [14, 15]. Notably, the JAK/STAT signaling pathway has been reported to be very important in hematological diseases [16] and many solid tumors, such as cervical cancer [17], breast cancer [18], hepatocellular carcinoma [19, 20], and acute myeloid leukemia (AML) [21]. Therefore, we proposed the hypothesis that exosomes might promote the transformation of NFs into CAFs depending on LINC00691 via JAK / STAT signaling pathway.

Our previous research results showed that exosomes contained in GC cell culture supernatants carry lncRNAs into cells and cause transformation in biological function [22]. The tumor microenvironment is composed of microenvironmental cells and an extracellular matrix, which plays crucial regulatory roles in almost all kinds of tumors [23]. CAFs are the vital stromal components that promote tumor progression in various ways [24]. In addition, the normal stroma of non-tumor tissues also contains a number of normal fibroblasts cells, which may be educated into CAFs. The high expression of α-smooth muscle actin (α-SMA) in CAFs is an important sign of activated fibroblasts [25].

This study aimed to estimate the clinical value of exosomal LINC00691 in the diagnosis assessment of GC and explore the mechanism of promoting CAFs formation through the JAK2/STAT3 signaling pathway.

Materials and methods

Patients and serum samples

A total of 94 serum samples were obtained from GC patients who were diagnosed with primary GC confirmed by pathological diagnosis in Nanjing Medical University Affiliated Suzhou Hospital from April 2019 to March 2022. All patients had not received surgical treatment, radiation therapy, chemotherapy or other adjuvant therapy before enrollment and had no other primary malignant tumors of digestive system or other systems. We also collected serum samples from 17 patients with benign gastric diseases including gastritis, gastric ulcer, and benign tumors diagnosed by endoscopy and pathology. In addition, 21 serum samples were obtained from healthy volunteers as control subjects. We have gotten informed consent from all of the mentioned patients and volunteers above. Also, this study was carried out after approval by the Institutional Ethical Committee of Nanjing Medical University on 2022 − 02–23. The number of the Ethics Committee decision is No. (2022)169.

Cell lines and cell culture

Human GC cell line HGC-27 and MKN-45(Guangzhou Cellcook Biotech Co., Ltd., Guangzhou, China) were cultured in complete medium, which contains 90% RPMI 1640 medium (Invitrogen, Carlsbad, CA) and 10% fetal bovine serum (FBS) in a CO2 incubator at 37 °C. Primary NFs and CAFs were collected from tumor tissues and paired normal samples. Briefly, the tissue was washed 3 times with PBS, soaked in 10,000 U of penicillin and streptomycin for 15 min, and cut into small pieces of 4 mm 2. And then 20% FBS-containing fibroblast medium (ScienCell Research Laboratories, San Diego, CA) was added for culture until the fibroblasts around the tissue block were confluent, and then they were digested by trypsin and passaged. In this study, we used NFs and CAFs of passages 5–8.

Exosome extraction

To exclude the effect of exosomes in serum, we centrifuged at 100,000 g for 18 h before extracting exosomes in the cell culture supernatant. The cell culture supernatant was collected and filtered using an exosome enrichment column (Merck Millipore Ltd. Darmstadt, Germany). Exosomes were then extracted using exosome extraction reagents (System Bioscience, LLC Palo Alto, CA) according to the protocol provided by the manufacturer.

RNA extraction and quantitative real-time polymerase chain reaction (qRT-PCR)

Total RNA was extracted via TRIzol reagent (Invitrogen) and then a HiFiScript cDNA Synthesis Kit (Cwbio, Beijing, China) was used to produce cDNA. Subsequently, RealSYBR Mixture (Cwbio) was applied in real-time qPCR performed in a real-time PCR Detection System (LightCycler 480, Roche, Switzerland). U6 was used as an internal reference. The sequences of primers used in this study were as follows: LINC00691-F: 5’-GCTCCCGATAAGCAACTGGA-3’; LINC00691-R: 5’-ACTCGTAGCCCAGAATCCCT-3’; U6-F: 5’-CTCGCTTCGGCAGCACA-3’; U6-R: 5’- AACGCTTCACGAATTTGCGT-3’.

Transmission electron microscopy (TEM)

Transmission electron microscopy was applied for exosome morphology. Briefly, 1% glutaraldehyde was used to fix exosomes in PBS and then a drop (20 μl) of exosomes was placed on 200-mesh Formvar/carbon-coated nickel grids, which were stained with 3% (W/V) aqueous phospphotungstic aid. The morphology of exosomes was recorded by transmission electron microscopy (Tecnai 12; Philips).

Nanoparticle size analysis

The exosome solution was diluted 10,000 times before being loaded on the nanoparticle size analyzer (#NanoSight NS300, Malven, England). It was carried out in accordance with the operating procedures provided by the manufacturer.

Immunofluorescence

Cells were fixed with 4% paraformaldehyde for 15 min and washed three times with PBS for 5 min each. Subsequently, cells were blocked with 0.3% Triton™ X-100 buffer for 60 min at room temperature. After blocking, the primary antibody was added and incubated overnight at 4 °C. Then, rinse three times with PBS for 5 min each before adding secondary antibody and incubating specimens for 1 h in the dark. Before photographing with a fluorescence microscope, three washes with PBS were performed for 5 min each.

Treatment of supernatant of cell culture medium

After fresh cell culture supernatant was collected, cell debris was removed by centrifugation at 1500 g for 10 min. When the cells were treated, an equal volume of the complete medium of the treated cells was added, and the cells were cultured at 37 °C with a 5% CO2 atmosphere for 48 h.

Cell transfection

For relevant materials and procedures of cell transfection, please refer to our previous article [10].

Mainly, GC cells were seeded into a 6-well cell culture plate (2 × 105 per well) and cultured overnight in 5% CO2 with antibiotic-free complete medium. LipoFiter transfection reagent (6 μl per well, Hanbio, Shanghai, China) was used to achieve the transfection of shRNAs targeting LINC00691 (1 μg per well, Hanbio) into GC cells in 1 ml serum-free RPMI-1640. The medium was then changed to complete medium containing 10% fetal bovine serum after transfection for 6 h. Subsequent experiments were performed at 36 h after transfection.

Cell counting assay and cell colony formation assay

After a 36 h transfection, 5 × 10 4 cells were seeded in 5 wells equally in 24-well plates. The total cell number was then counted every 24 h. GraphPad 5.0 was applied to draw growth curves depending on cell number. Also, cells were seeded in 6-well plates for colony formation assay (1 × 104 per well) and cultured t 37 °C with a 5% CO2 atmosphere for 7 days. Subsequently, cells were fixed using 4% paraformaldehyde and then stained with crystal violet. The colony number was finally calculated using a microscope.

Transwell migration and invasion assay

After a 36 h transfection, 2 × 104 cells were seeded into the top chamber of Transwell chambers (Chemicon, Temecula, CA, USA) resuspended in a 200 μl serum-free medium. Complete medium (600 μl) was added into the bottom chamber. For invasion assay, there was an increased step compared to the migration experiment: the top chamber was added with 20% Matrigel matrix 8 h before seeding cells and the number of seeded cells was 1 × 105. After migration or invasion for 36 h, cells were fixed using 4% paraformaldehyde for 30 min at room temperature, stained with crystal violet for 15 min, and counted under the microscope after the upper layer cells were carefully removed.

Wound healing assay

A total of 2 × 104 cells were seeded in a 24 well plate overnight, and then a straight line was drawn in the middle of every well with a 10 μl pipette tip. After being washed three times with PBS, cells were changed to a serum-free medium. Cells were then photographed and recorded after 0, 4, and 8 h in a CO2 incubator at 37 °C.

Western blot analysis

RIPA buffer (Beyotime, Shanghai, China) was mixed with 0.1% protease inhibitor PMSF (Beyotime) and applied to lyse cells. The protein samples were denatured by heating and mixed with loading buffer before electrophoresis. SDS–polyacrylamide gel electrophoresis (SDS-PAGE) was performed to separate proteins. Then target proteins were electrophoretically transferred at 360 mA current for 120 min and collected on 0.22-μm polyvinylidene fluoride (PVDF) membranes (Millipore, MA, USA). 5% skim milk was used to block non-specific binding sites for 60 min at room temperature. PVDF membranes were then cropped prepared for applying different primary antibody. Primary antibody was applied to bind the target protein on PVDF membranes overnight at 4 °C. The membrane was washed three times for 5 min each with TBS/T and incubated with the secondary antibody for 60 min at 37 °C. Scientific SuperSignal chemiluminescent HRP substrates (Thermo Fisher, Waltham, USA) was then applied matched with an enhanced chemiluminescence kit according to the operating procedures provided by the manufacturer. GAPDH was applied as an internal reference.

Statistical analysis

Mainly, we used SPSS 22.0 (Chicago, IL, USA) for statistical analyses and GraphPad Prime 5 (GraphPad, San Diego, CA) for statistical charts. All experiments mentioned in this study were repeated thrice at least. Values recorded in this study are performed as mean ± SD. Student’s t-test was applied to analyze the statistical difference between paired groups. ANOVA and Newman-Keuls Multiple Comparison Test was used to test whether there was any statistical difference among the three groups of treated samples. Pearson χ2 test was used to analyze if there was any correlation between serum exosomal LINC00691 level and clinicopathological features of GC patients. P < 0.05 was considered as a statistically significant difference.

Results

Exosomal LINC00691 level in serum of GC patients was higher than that of healthy people

We extracted serum exosomes from 94 GC patients and detected the expression of LINC00691. The results indicated that the LINC00691 level in serum exosomes of GC patients was significantly higher than that in healthy people and patients with gastric benign disease (Fig. 1a). Moreover, the results of pathological correlation analysis showed that the level of serum exosomal LINC00691 was associated with tumor size, lymph node metastasis, vascular invasion and perineural invasion of patients (Table 1). The area under the curve (AUC) was 0.86, which was higher than that of serum LINC00691. The sensitivity was 0.85 and the specificity was 0.71 respectively (Fig. 1b). These results demonstrated that LINC00691 in serum exosomes may be an ideal biomarker for GC. Besides, we collected a part of GC patients' serum after gastrectomy and detected the expression of exosomal LINC00691. The results manifested that the exosomal LINC00691 level in the serum of GC patients was significantly reduced after gastrectomy (Fig. 1c).

Fig. 1
figure 1

Exosomal LINC00691 level in serum of GC patients, patients with benign gastric diseases, and healthy people. a Scatter plot of serum exosomal LINC00691 levels in patients with GC (n = 94), benign gastric diseases (n = 17), and healthy people (n = 21). The expression levels of Exosomal LINC00691 were determined by using qRT-PCR (***P < 0.001). Data were presented as mean ± SD. b The ROC curve of serum exosomal LINC00691 in GC. c Changes of serum exosomal LINC00691 levels in GC patients before and after GC resection (**P < 0.01)

Table 1 The association between serum exosomal LINC00691 expression levels (–ΔCt) and the clinicopathological features of gastric cancer patients

Isolation and identification of primary NFs and CAFs

We isolated and cultured CAFs and NFs from GC tumor tissues and paired healthy tissues. We observed microscopically similar morphologies of NFs and CAFs, but the former was a sinner and more elongated with relatively fewer cell processes (Fig. 2a). We collected CAFs and NFs in generations 3–8 and found the expression of LINC00691 in CAFs (4.58 ± 0.99) was higher than that in NFs (1.33 ± 0.33) (Fig. 2b). Since α-SMA and Vimentin have been reported as activation markers of CAFs, we examined their distribution in NFs and CAFs by immunofluorescence. The results indicated that the expression of both α-SMA and Vimentin in CAFs was higher than NFs (Fig. 2c).

Fig. 2
figure 2

Isolation and identification of primary NFs and CAFs. a The morphology of NFs and CAFs under the microscope. Magnification, × 200. Scale bar, 20 μm. b The expression levels of LINC00691 in NFs and CAFs were determined by using qRT-PCR (*P < 0.05). c Immunofluorescence staining of α-SMA and Vimentin in NFs and CAFs. Magnification, × 200. Scale bar, 20 µm

Treatment of NFs with exosomes from GC cells enhances the cell viability of NFs

To explore the effects of exosomes from GC cells on NFs, we isolated and detected exosomes from GC cells. It was found that exosomes in GC cell supernatants were spherical vesicles of 40–150 nm in size using transmission electron microscopy (TEM) (Fig. 3a). Western blot results showed that purified exosomes expressed exosomal biomarkers CD9, CD81, CD63, and Alix (Fig. 3b). we also used NTA to analyze the size distribution and concentration of exosomes. Approximately, isolated exosomes were mainly distributed in diameters ranging from 50 to 200 nm in size and the concentration ranged from 1.0 × 10 11 to 2.5 × 10 10 particles/ml (Fig. 3c). Exosomes observed by NTA showed similar morphology to that under electron microscope (Fig. 3d).

Fig. 3
figure 3

Identification of exosomes. a Morphology of exosomes photographed by transmission electron microscopy (TEM). Scale bar, 100 nm. b Western blot detection of exosome marker molecules CD9, CD81, CD63, and Alix. c Nanoparticle size analysis of the particle size/concentration of exosomes from HGC-27 cells. d Morphology of exosomes photographed by (Nanoparticle Tracking Analysis, NTA)

We treated NFs with exosomes from GC cell culture supernatant and found that the migration of NFs was enhanced through wound healing assay and Transwell migration assay, which was significantly inhibited when NFs were treated with exosome-depleted GC cell culture supernatants (Fig. 4a, b). Plate cloning experiments showed that exosomes derived from GC cell culture supernatant promoted NFs to acquire stronger clonogenic ability. Nevertheless, this effect was distinctly suppressed when exosomes were removed by centrifugation (Fig. 4c). Similarly, growth curve assay indicated that GC cell culture supernatant-derived exosomes promoted NFs to acquire stronger proliferative capacity (Fig. 4d). Besides, exosomes derived from GC cell culture supernatant enhanced the expression of LINC00691in NFs (P < 0.05, compared to ctrl, Fig. 4e).

Fig. 4
figure 4

NFs treated with exosomes from GC cell culture supernatant. a Wound healing assay of fibroblasts after being treated with exosomes. Magnification, × 40. Scale bar, 100 µm. b Transwell migration assay of fibroblasts after being treated with exosomes. Magnification, × 40. Scale bar, 100 µm. c Cell colony formation assay of fibroblasts after being treated with exosomes. Magnification, × 100. Scale bar, 20 µm. d Cell counting assay of fibroblasts after treated with exosomes. e The expression levels of LINC00691of fibroblasts after being treated with exosomes (P < 0.05, compared to ctrl)

GC cell culture supernatant treatment replicated the effects of exosome treatment

GC cell culture supernatant was applied to treat NFs and it was found that the expression of α-SMA and Vimentin in NFs was significantly increased (Fig. 5a). It has been reported that activated fibroblasts enhanced the proliferation and invasion of GC cells [1]. We subsequently treated GC cells with culture supernatant derived from the treated fibroblasts. The proliferation of GC cells was significantly enhanced when treated with fibroblasts pretreated with GC cell culture supernatants (Fig. 5b). Clonogenicity was likewise promoted by fibroblasts pretreated with GC cell culture supernatants (Fig. 5c). Transwell migration and invasion assay manifested that GC cells acquired reinforced migration and invasion abilities benefited from fibroblasts activated by GC cell culture supernatants (Fig. 5d).

Fig. 5
figure 5

Biological effects of GC supernatants containing exosomes on NFs. a Immunofluorescence staining of α-SMA and Vimentin in fibroblasts after being treated with GC supernatants. Magnification, × 200. Scale bar, 20 µm. b Cell counting assay of GC cells after being treated with fibroblasts induced by supernatants. c Cell colony formation assay of GC cells after being treated with fibroblasts induced by supernatants. d Transwell migration and invasion assay of GC cells after being treated with fibroblasts induced by supernatants. Magnification, × 200. Scale bar, 10 µm, **P < 0.01

Knockdown of LINC00691 suppressed the effects of GC cell culture supernatant on NFs

As we found the expression of LINC00691 of NFs was upregulated when treated with exosomes derived from GC cell culture supernatant (Fig. 4e), we then knockdown LINC00691 in GC cells and collected culture supernatant. The results indicated that the inhibition of LINC00691 led to the restraints of effects of GC cell culture supernatant on NFs. Specifically, the expression of fibroblast activated biomarkers α-SMA and Vimentin was significantly decreased (Fig. 6a). The proliferation, clonogenicity, migration, and invasion of GC cells were weakened when treated by pretreated fibroblasts compared with the control group (Fig. 6b-d).

Fig. 6
figure 6

Biological effects of LINC00691 in GC supernatants on NFs. a Immunofluorescence staining of α-SMA and Vimentin in fibroblasts after being treated with LINC00691 knockdown GC supernatants. Magnification, × 200. Scale bar, 20 µm. b Cell counting assay of GC cells after being treated with fibroblasts induced by LINC00691 knockdown supernatants. c Cell colony formation assay of GC cells after being treated with fibroblasts induced by LINC00691 knockdown supernatants. d Transwell migration and invasion assay of GC cells after being treated with fibroblasts induced by LINC00691 knockdown supernatants. Magnification, × 200. Scale bar, 10 µm, *P < 0.05, ***P < 0.001

The inhibition of JAK2/STAT3 signaling pathway suppressed the effects of GC cell culture supernatant on NFs

In previous studies, we elucidated that LINC00691 promoted GC malignant progression through the JAK2/STAT3 signaling pathway. Therefore, we use the inhibitor of the JAK2/STAT3 signaling pathway called ruxolitinib in the system when we applied GC cell culture supernatant to treat NFs. It was found that the expression of fibroblast activated biomarkers α-SMA and Vimentin was apparently reduced by ruxolitinib (Fig. 7a). Afterwards, we treated GC cells with culture supernatant derived from the treated fibroblasts. The results indicated that the effects of induced fibroblasts of promoting proliferation, clonogenicity, migration, and invasion on GC cells activated by GC cell culture supernatants were remarkably suppressed compared with the control group (Fig. 7b-d).

Fig. 7
figure 7

Biological effects of JAK2/STAT3 signaling pathway in GC supernatants on NFs. a Immunofluorescence staining of α-SMA and Vimentin in fibroblasts after being treated with ruxolitinib pre-treated GC supernatants. Magnification, × 200. Scale bar, 20 µm. b Cell counting assay of GC cells after being treated with fibroblasts induced by ruxolitinib pre-treated supernatants. c Cell colony formation assay of GC cells after being treated with fibroblasts induced by ruxolitinib pre-treated supernatants. d Transwell migration and invasion assay of GC cells after being treated with fibroblasts induced by ruxolitinib pre-treated supernatants. Magnification, × 200. Scale bar, 10 µm, **P < 0.01, ***P < 0.001

Discussion

It is widely reported that lncRNAs play indispensable roles in various cell life process, including but not limited to cell differentiation, proliferation, migration, apoptosis and senescence [26]. In particular, a large number of studies in recent years have illuminated the potential applications of lncRNAs as biomarkers or therapeutic targets in cancer progression and treatment tolerance [27,28,29]. In the previous study, we reported that overexpressed LINC00691 in GC tissues and serum was a potential biomarker for GC, which was correlated with tumor size, depth of invasion, lymph node metastasis, venous invasion, perineural invasion, and poor prognosis of patients [10]. We reported that LINC00691 promoted the proliferation and invasion of GC cells by activating Lin28 transcription and facilitating EGF expression through the JAK2/STAT3 signalling pathway. Other related studies reported that LINC00691 was abnormally expressed in non-small cell lung cancer, lung adenocarcinoma, osteosarcoma, and papillary thyroid cancer [30,31,32,33]. However, the functions of exosomal LINC00691 in GC remain unknown. In this study, we reported that LINC00691 expression in serum exosomes of GC patients was significantly higher than that of healthy volunteers and patients with gastric benign disease and it was associated with tumor size, lymph node metastasis, vascular invasion and perineural invasion of patients. The ROC curve showed that the sensitivity and specificity of diagnosis were: 0.85 and 0.71 respectively. However, limitations of using exosomal LINC00691 as a diagnosis biomarker for GC also exists. This result was based on a small local population, and the sample number of healthy volunteers in the control group was also small. More precise studies are needed in a larger population.

Increasing number of reports have indicated that tumor cell-derived exosomes play a prominent educational role in the tumor microenvironment [34]. Recent studies reported exosomes used as special drug delivery systems for targeted therapy reprogramming tumor microenvironment [35]. As important components of the microenvironment, fibroblasts may be involved in the activation of early oncogenic signals and regulate tumor progression after activation [36]. CAFs, considered as a set of activated fibroblasts produce extracellular matrix components such as cytokines, growth factors, and chemokines to support the growth and invasion of cancer cells [37]. CAFs are usually identified by detecting markers such as α-SMA, Vimentin, and fibroblast activation protein alpha (FAP) [38, 39]. We isolated CAFs and NFs from GC tissues and paired healthy tissues, and found that the expression levels of α-SMA and Vimentin were significantly higher in CAFs than that in NFs. After treatment with GC cell culture supernatant or exosomes, the expression of α-SMA and Vimentin in fibroblasts was significantly increased, which proved that fibroblasts were activated. Then the activation of fibroblasts was inhibited as exosomes in the supernatant of GC cells were deprived. Our results are consistent with the study reported [40, 41].

It was reported that conditioned media of CAFs promoted cancer calls to gain a more proliferative phenotype [42]. We threated GC cells with the culture supernatant of activated fibroblasts induced by exosomes and found that not only the proliferation of GC cells was enhanced, but also the migration and invasion were boosted. Radiotherapy to CAFs induced insulin-like growth factor-1 (IGF1) and stimulated survival of cancer cells, which was restrained by IGF1 receptor (IGF1R) neutralization [43]. This may be a potential therapeutic strategy targeting key soluble factors secreted by fibroblasts. LncRNA H19 was overexpressed in tumor tissues and facilitated the stemness and chemoresistance of colorectal cancer (CRC) through the regulation of CAFs [44]. We manifested that CAFs as well as activated fibroblasts induced by exosomes derived from GC cell culture supernatant expressed a higher level of LINC00691. Sh-RNAs targeted LINC00691 inhibited the effects of GC cell culture supernatant on fibroblasts. This study manifested that exosomes of GC cells promoted normal fibroblasts to gain more activated phenotypes similar to CAFs depending on LINC00691 partly at least. However, there are still more details about the underlying molecular mechanisms of exosomes-rich GC cell culture supernatant on normal fibroblasts needed clarified. Moreover, inhibitor of the JAK2/STAT3 signaling pathway ruxolitinib also suppressed the activation of normal fibroblasts. Biffi et al. reported that JAK/STAT activation was a dominant factor that promoted an inflammatory CAF state in pancreatic ductal adenocarcinoma (PDAC) [45]. Ruxolitinib is currently clinically applied for the treatment of patients with intermediate or high-risk myelofibrosis including primary myelofibrosis, post-polycythemia vera myelofibrosis and post-essential thrombocythemia myelofibrosis [46, 47]. In recent years, some studies have indicated that ruxolitinib may have potential applications in targeted therapy of other diseases, such as renal interstitial fibrosis [48], graft-versus-host disease [49] and severe coronavirus disease 2019 (COVID-19) [50].

Mainly, CAFs in GC were identified into two subsets, inflammatory CAFs (iCAFs) and extracellular matrix CAFs (eCAFs) by performing single-cell RNA sequencing [51]. Another study revealed myofibroblastic CAF (myCAF) and inflammatory CAF (iCAF) when tumors metastasized to the liver [52]. Although different CAF subtypes and classification are illustrated to have different biological functions, they are usually manifested to coordinate the tumor microenvironment to promote cancer progression [38]. In this study, we did not identify the subtype or classification of fibroblasts activated by exosomes in GC cell culture supernatants, which would be further explored in subsequent studies. Our study revealed that exosomal LINC00691 promoted NFs to obtain activated phenotype like CAFs through JAK/STAT signaling pathways. This study uncovered new therapeutic application in GC. However, there are more efforts needed to be put into practice before the clinical therapeutic application of ruxolitinib in GC. We need to verify the role of ruxolitinib in GC therapy in vivo in subsequent studies. Still there is no any clinical study having indicated that ruxolitinib benefits GC patients with a longer survival time.

Overall, our study indicated that serum exosomal LINC00691 was a potential diagnosis biomarker for GC. Exosomes derived from GC cell culture supernatant stimulated the activation of normal fibroblasts to gain a CAF-like phenotype, which performed a higher level of α-SMA and Vimentin and promoted the proliferation and invasion of GC cells.

Availability of data and materials

All data generated or analyzed during this study are included in this published article (and its supplementary information files).

Abbreviations

GC:

Gastric cancer

NFs:

Normal fibroblasts

CAFs:

cancer-associated fibroblasts

JAK2/STAT3:

Janus kinase 2 /signal transducer and activator of transcription 3

References

  1. Grunberg N, Pevsner-Fischer M, Goshen-Lago T, Diment J, Stein Y, Lavon H, Mayer S, Levi-Galibov O, Friedman G, Ofir-Birin Y, et al. Cancer-associated fibroblasts promote aggressive gastric cancer phenotypes via heat shock factor 1-mediated secretion of extracellular vesicles. Cancer Res. 2021;81(7):1639–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Sun K, Lv H, Chen B, Nie C, Zhao J, Wang S, Wang J, Xu W, Chen X. Dawning precision treatment for gastric cancer: The latest biomarkers. J Transl Int Med. 2021;9(4):228–30.

    PubMed  PubMed Central  Google Scholar 

  3. So JBY, Kapoor R, Zhu F, Koh C, Zhou L, Zou R, Tang YC, Goo PCK, Rha SY, Chung HC, et al. Development and validation of a serum microRNA biomarker panel for detecting gastric cancer in a high-risk population. Gut. 2021, 70(5):829–837.

  4. Li G, Wang G, Chi F, Jia Y, Wang X, Mu Q, et al. Higher postoperative plasma EV PD-L1 predicts poor survival in patients with gastric cancer. J Immunother Cancer. 2021;9(3):e002218. https://doi.org/10.1136/jitc-2020-002218.

  5. Li P, Zhang Y, Xu Y, Cao H, Li L. Characteristics of CD8+ and CD4+ tissue-resident memory lymphocytes in the gastrointestinal tract. Advanced Gut & Microbiome Research. 2022;2022:9157455.

    Google Scholar 

  6. Wolfe W, Xiang Z, Yu X, Li P, Chen H, Yao M, Fei Y, Huang Y, Yin Y, Xiao H. The challenge of applications of probiotics in gastrointestinal diseases. Adv Gut Microbiome Res. 2023;2023:1984200.

    Google Scholar 

  7. Kopp F, Mendell JT. Functional Classification and Experimental Dissection of Long Noncoding RNAs. Cell. 2018;172(3):393–407.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Venkatesh J, Wasson MD, Brown JM, Fernando W, Marcato P. LncRNA-miRNA axes in breast cancer: Novel points of interaction for strategic attack. Cancer Lett. 2021;509:81–8.

    CAS  PubMed  Google Scholar 

  9. Wu P, Mo Y, Peng M, Tang T, Zhong Y, Deng X, Xiong F, Guo C, Wu X, Li Y, et al. Emerging role of tumor-related functional peptides encoded by lncRNA and circRNA. Mol Cancer. 2020;19(1):22.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Liang W, Xia B, He C, Zhai G, Li M, Zhou J. Overexpression of LINC00691 promotes the proliferation and invasion of gastric cancer cells via the Janus kinase/signal transducer and activator of transcription signalling pathway. Int J Biochem Cell Biol. 2020;123: 105751.

    CAS  PubMed  Google Scholar 

  11. Wang L, Wu J, Song S, Chen H, Hu Y, Xu B, Liu J. Plasma exosome-derived sentrin SUMO-specific protease 1: a prognostic Biomarker in patients with osteosarcoma. Front Oncol. 2021;11.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. An Y, Lin S, Tan X, Zhu S, Nie F, Zhen Y, Gu L, Zhang C, Wang B, Wei W, et al. Exosomes from adipose-derived stem cells and application to skin wound healing. Cell Prolif. 2021;54(3): e12993.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Fan J, Xu G, Chang Z, Zhu L, Yao J. miR-210 transferred by lung cancer cell-derived exosomes may act as proangiogenic factor in cancer-associated fibroblasts by modulating JAK2/STAT3 pathway. Clin Sci (Lond). 2020;134(7):807–25.

    CAS  PubMed  Google Scholar 

  14. Montero P, Milara J, Roger I, Cortijo J. Role of JAK/STAT in Interstitial Lung Diseases; Molecular and Cellular Mechanisms. Int J Mol Sci. 2021;22(12):6211. https://doi.org/10.3390/ijms22126211.

  15. Darnell JE Jr, Kerr IM, Stark GR. Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 1994;264(5164):1415–21.

    CAS  PubMed  Google Scholar 

  16. Wu J, Lu WY, Cui LL. Clinical significance of STAT3 and MAPK phosphorylation, and the protein expression of cyclin D1 in skin squamous cell carcinoma tissues. Mol Med Rep. 2015;12(6):8129–34.

    CAS  PubMed  Google Scholar 

  17. Gutiérrez-Hoya A, Soto-Cruz I. Role of the JAK/STAT Pathway in Cervical Cancer: its Relationship with HPV E6/E7 Oncoproteins. Cells. 2020;9(10):2297. https://doi.org/10.3390/cells9102297.

  18. Shao F, Pang X, Baeg GH. Targeting the JAK/STAT Signaling Pathway for Breast Cancer. Curr Med Chem. 2021;28(25):5137–51.

    CAS  PubMed  Google Scholar 

  19. Toh TB, Lim JJ, Hooi L, Rashid M, Chow EK. Targeting Jak/Stat pathway as a therapeutic strategy against SP/CD44+ tumorigenic cells in Akt/beta-catenin-driven hepatocellular carcinoma. J Hepatol. 2020;72(1):104–18.

    CAS  PubMed  Google Scholar 

  20. Liu P, Zhu H, Zhu H, Zhang X, Feng A, Zhu X, Sun Y. Predicting survival for hepatic arterial infusion chemotherapy of unresectable colorectal liver metastases: radiomics analysis of pretreatment computed tomography. J Transl Int Med. 2022;10(1):56–64.

    PubMed  PubMed Central  Google Scholar 

  21. Chen BR, Deshpande A, Barbosa K, Kleppe M, Lei X, Yeddula N, Vela PS, Campos AR, Wechsler-Reya RJ, Bagchi A, et al. A JAK/STAT-mediated inflammatory signaling cascade drives oncogenesis in AF10-rearranged AML. Blood. 2021;137(24):3403–15.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Pan L, Liang W, Fu M, Huang ZH, Li X, Zhang W, Zhang P, Qian H, Jiang PC, Xu WR, et al. Exosomes-mediated transfer of long noncoding RNA ZFAS1 promotes gastric cancer progression. J Cancer Res Clin Oncol. 2017;143(6):991–1004.

    CAS  PubMed  Google Scholar 

  23. Pine AR, Cirigliano SM, Nicholson JG, Hu Y, Linkous A, Miyaguchi K, Edwards L, Singhania R, Schwartz TH, Ramakrishna R, et al. Tumor microenvironment is critical for the maintenance of cellular states found in primary glioblastomas. Cancer Discov. 2020;10(7):964–79.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Oya Y, Hayakawa Y, Koike K. Tumor microenvironment in gastric cancers. Cancer Sci. 2020;111(8):2696–707.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Díaz-Maroto NG, Garcia-Vicién G, Polcaro G, Bañuls M, Albert N, Villanueva A, Molleví DG. The blockade of tumoral IL1β-mediated signaling in normal colonic fibroblasts sensitizes tumor cells to chemotherapy and prevents inflammatory CAF activation. Int J Mol Sci. 2021;22(9):4960. https://doi.org/10.3390/ijms22094960.

  26. McCabe EM, Rasmussen TP. lncRNA involvement in cancer stem cell function and epithelial-mesenchymal transitions. Semin Cancer Biol. 2021;75:38–48.

    CAS  PubMed  Google Scholar 

  27. Liu SJ, Dang HX, Lim DA, Feng FY, Maher CA. Long noncoding RNAs in cancer metastasis. Nat Rev Cancer. 2021;21(7):446–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Malla RR. Long noncoding RNAs: potential mediators of liver cancer metastasis. Crit Rev Oncog. 2021;26(1):21–33.

    PubMed  Google Scholar 

  29. Yousefi H, Maheronnaghsh M, Molaei F, Mashouri L, Reza Aref A, Momeny M, Alahari SK. Long noncoding RNAs and exosomal lncRNAs: classification, and mechanisms in breast cancer metastasis and drug resistance. Oncogene. 2020;39(5):953–74.

    CAS  PubMed  Google Scholar 

  30. Li Z, Gao Y, Li L, Xie S. Curcumin inhibits papillary thyroid cancer cell proliferation by regulating lncRNA LINC00691. Anal Cell Pathol (Amst). 2022;2022:5946670.

    PubMed  Google Scholar 

  31. Xie Y, Hu X. Increased levels of long noncoding RNA LINC00691 correlate with poor prognosis in non-small-cell lung cancer patients. J Clin Lab Anal. 2020;34(8): e23357.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhao B, Xu H, Ai X, Adalat Y, Tong Y, Zhang J, Yang S. Expression profiles of long noncoding RNAs in lung adenocarcinoma. Onco Targets Ther. 2018;11:5383–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Wan D, Qu Y, Zhang L, Ai S, Cheng L. The lncRNA LINC00691Functions as a ceRNA for miRNA-1256 to Suppress Osteosarcoma by Regulating the Expression of ST5. Onco Targets Ther. 2020;13:13171–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Meng W, Hao Y, He C, Li L, Zhu G. Exosome-orchestrated hypoxic tumor microenvironment. Mol Cancer. 2019;18(1):57.

    PubMed  PubMed Central  Google Scholar 

  35. Zhou W, Zhou Y, Chen X, Ning T, Chen H, Guo Q, Zhang Y, Liu P, Zhang Y, Li C, et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials. 2021;268: 120546.

    CAS  PubMed  Google Scholar 

  36. Helms E, Onate MK, Sherman MH. Fibroblast Heterogeneity in the Pancreatic Tumor Microenvironment. Cancer Discov. 2020;10(5):648–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Elyada E, Bolisetty M, Laise P, Flynn WF, Courtois ET, Burkhart RA, Teinor JA, Belleau P, Biffi G, Lucito MS, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals antigen-presenting cancer-associated fibroblasts. Cancer Discov. 2019;9(8):1102–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Nurmik M, Ullmann P, Rodriguez F, Haan S, Letellier E. In search of definitions: Cancer-associated fibroblasts and their markers. Int J Cancer. 2020;146(4):895–905.

    CAS  PubMed  Google Scholar 

  39. Richards KE, Zeleniak AE, Fishel ML, Wu J, Littlepage LE, Hill R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene. 2017;36(13):1770–8.

    CAS  PubMed  Google Scholar 

  40. Liu J, Ren L, Li S, Li W, Zheng X, Yang Y, Fu W, Yi J, Wang J, Du G. The biology, function, and applications of exosomes in cancer. Acta Pharm Sin B. 2021;11(9):2783–97.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Baroni S, Romero-Cordoba S, Plantamura I, Dugo M, D’Ippolito E, Cataldo A, Cosentino G, Angeloni V, Rossini A, Daidone MG, et al. Exosome-mediated delivery of miR-9 induces cancer-associated fibroblast-like properties in human breast fibroblasts. Cell Death Dis. 2016;7(7): e2312.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Suh J, Kim DH, Lee YH, Jang JH, Surh YJ. Fibroblast growth factor-2, derived from cancer-associated fibroblasts, stimulates growth and progression of human breast cancer cells via FGFR1 signaling. Mol Carcinog. 2020;59(9):1028–40.

    CAS  PubMed  Google Scholar 

  43. Tommelein J, De Vlieghere E, Verset L, Melsens E, Leenders J, Descamps B, Debucquoy A, Vanhove C, Pauwels P, Gespach CP, et al. Radiotherapy-activated cancer-associated fibroblasts promote tumor progression through paracrine IGF1R activation. Cancer Res. 2018;78(3):659–70.

    CAS  PubMed  Google Scholar 

  44. Ren J, Ding L, Zhang D, Shi G, Xu Q, Shen S, Wang Y, Wang T, Hou Y. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018;8(14):3932–48.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Biffi G, Oni TE, Spielman B, Hao Y, Elyada E, Park Y, Preall J, Tuveson DA. IL1-Induced JAK/STAT signaling is antagonized by TGFbeta to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov. 2019;9(2):282–301.

    PubMed  Google Scholar 

  46. Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger M, Miller C, Silver RT, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366(9):799–807.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Vannucchi AM, Kiladjian JJ, Griesshammer M, Masszi T, Durrant S, Passamonti F, Harrison CN, Pane F, Zachee P, Mesa R, et al. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med. 2015;372(5):426–35.

    PubMed  PubMed Central  Google Scholar 

  48. Bai Y, Wang W, Yin P, Gao J, Na L, Sun Y, Wang Z, Zhang Z, Zhao C. Ruxolitinib Alleviates Renal Interstitial Fibrosis in UUO Mice. Int J Biol Sci. 2020;16(2):194–203.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Ali H, Salhotra A, Modi B, Nakamura R. Ruxolitinib for the treatment of graft-versus-host disease. Expert Rev Clin Immunol. 2020;16(4):347–59.

    CAS  PubMed  Google Scholar 

  50. Cao Y, Wei J, Zou L, Jiang T, Wang G, Chen L, Huang L, Meng F, Huang L, Wang N, et al. Ruxolitinib in treatment of severe coronavirus disease 2019 (COVID-19): a multicenter, single-blind, randomized controlled trial. J Allergy Clin Immunol. 2020; 146(1):137–146 e133.

  51. Li X, Sun Z, Peng G, Xiao Y, Guo J, Wu B, Li X, Zhou W, Li J, Li Z, et al. Single-cell RNA sequencing reveals a pro-invasive cancer-associated fibroblast subgroup associated with poor clinical outcomes in patients with gastric cancer. Theranostics. 2022;12(2):620–38.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Bhattacharjee S, Hamberger F, Ravichandra A, Miller M, Nair A, Affo S, et al. Tumor restriction by type I collagen opposes tumor-promoting effects of cancer associated fibroblasts. J Clin Invest. 2021;131(11):e146987. https://doi.org/10.1172/JCI146987.

Download references

Acknowledgements

The authors would like to thank Dr. Xu Zhang (School of Medicine, Jiangsu University) for his kindly providing technical assistance and guidance in experiments.

Funding

This study was partially sponsored by Suzhou Project in Science and Education (grant no. kjxw2018035) and People's Livelihood Science and Technology Guiding Project (grant no. SKJYD2021106).

Author information

Authors and Affiliations

Authors

Contributions

Conception and design: Wei Liang and Chunrong Sun; Administrative support: Bin Xia and Chunrong Sun; Provision of study materials or patients: Bin Xia, Xiuyu Gu and Tingting Xu; Collection and assembly of data: Meina Yan Lan Huang, Chun Jiang and Meifen Li; Data analysis and interpretation: Guanghua Zhai, Guoping Zhang, Jian Wu and Yu Zhou; Manuscript writing: All authors; Final approval of manuscript: All authors.

Corresponding authors

Correspondence to Yu Zhou, Chunrong Sun or Wei Liang.

Ethics declarations

Ethics approval and consent to participate

The study was carried out after approval by the Institutional Ethical Committee of Nanjing Medical University on 2022 − 02–23. The number of the Ethics Committee decision is No. (2022)169. Consent of informed consent was confirmed by patients or their relatives for clinical samples used in this study. All methods were performed in accordance with the relevant guidelines and regulations or in accordance with the Declaration of Helsinki.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

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

Supplementary Information

Additional file 1: Table S1.

Clinicopathological features of gastric cancer patients. Alix and CD63 for Fig.3b. CD9 for Fig.3b. CD81 for Fig.3b. 

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xia, B., Gu, X., Xu, T. et al. Exosomes-mediated transfer of LINC00691 regulates the formation of CAFs and promotes the progression of gastric cancer. BMC Cancer 23, 928 (2023). https://doi.org/10.1186/s12885-023-11373-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-023-11373-5

Keywords