- Research
- Open access
- Published:
Intraovarian injection of 3D-MSC-EVs-ECM gel significantly improved rat ovarian function after chemotherapy
Reproductive Biology and Endocrinology volume 22, Article number: 125 (2024)
Abstract
Background
Restoring the function of the ovary is important for chemotherapy-induced ovarian failure (COF) patients. Stem cell and extracellular vesicles (EVs) therapy show promise but need further improvement.
Methods
Human umbilical cord mesenchymal stem cells (hUC-MSCs) were primarily cultured and further three-dimensional (3D) cultured using an ultra-low attachment surface method. The expression levels of nutritional cytokines and immunomodulatory and stemness-related genes of 3D-cultured hUC-MSCs were analyzed. EVs were isolated by ultracentrifugation and characterized. Ovaries were decellularized with sodium dodecyl sulfate to obtain extracellular matrix (ECM). Lyophilized EVs from three-dimensional (2D) or 3D hUC-MSCs were mixed with ECM to prepare the 2D/3D-MSC-EVs-ECM gels. The therapeutic effect of the MSC-EVs-ECM gel on cyclophosphamide (CTX) -treated rats was analyzed through various tests. RNA sequencing was used to analyze the expression changes of genes before and after treatment.
Results
After culturing in ultra-low attachment dishes, hUC-MSCs aggregated into spheroids and significantly upregulated the expression levels of immunomodulatory and stemness-related genes. The total EVs yield was also upregulated (5.6-fold) after 3D culture. The cell viability of CTX-treated ovarian granulosa cells (OGCs) was significantly rescued by coculture with the 3D-MSC-EVs-ECM gel. Hormones indicative of ovarian function, AMH, E2, and FSH, were recovered in both the CTX + 2D-MSC-EVs-ECM gel group and the CTX + 3D-MSC-EVs-ECM gel group, while the apoptosis-related protein Bax was significantly downregulated. The 3D-MSC-EVs-ECM gel was more effective than the 2D-MSC-EVs-ECM gel. Significantly differentially expressed genes, such as Hbb-b1, Gpd1, and Sirpa, were detected by RNA sequencing. Hbb-b1 was increased in the ovaries of CTX-treated rats, and this increase was attenuated by injecting the 2D/3D-MSC-EVs-ECM gel. Gpd1 was increased after CTX treatment, and this increase was reversed by the 3D-MSC-EVs-ECM gel. Sirpa was decreased in the ovaries of CTX-treated rats, and this decrease was attenuated by injecting the 3D-MSC-EVs-ECM gel.
Conclusions
Our study demonstrated that the 3D-MSC-EVs-ECM gel is an efficient strategy for the recovery of ovarian function in CTX-induced ovarian failure.
Introduction
In recent years, the burden of cancer has risen worldwide in adults, adolescents, and children [1, 2]. Current cancer treatments include chemotherapy, radiotherapy, surgery, and immunotherapy [3]. Chemotherapy is a common treatment option [4]. However, chemotherapy can cause many side effects, including myelosuppression, gastrointestinal reactions, and liver and kidney dysfunction. Among these adverse effects, the major concern of survivors and their parents are gonadal dysfunction and infertility [5]. Many chemotherapy drugs can cause ovarian failure, known as chemotherapy associated ovarian failure (COF) [6]. Cyclophosphamide (CTX) has a high risk of causing COF. CTX was the first chemotherapy drug to be associated with ovarian failure. It can induce ovarian granulosa apoptosis and follicular atresia and accelerate the activation of the primordial follicles [7, 8].
Currently, there are several available methods for ovarian protection from COF, such as embryo and oocyte cryopreservation, ovarian tissue cryopreservation, and gonadotrophin-releasing hormone analogs [6, 7, 9]. However, these methods have some limitations. For embryo and oocyte cryopreservation, oocytes need to be obtained through ovulation induction, which will delay tumor treatment and may increase the risk of invasion and metastasis of sex hormone-dependent tumors [6]. Also, this method cannot be performed on prepubertal girls. For ovarian tissue cryopreservation, the risk of tumor recurrence due to reintroduction of cancer cells by transplantation of cryopreserved ovarian tissue is unknown and more studies evaluating the safety of autologous ovarian transplantation in cancer patients should be needed in the future [6]. Besides, not all studies have confirmed that gonadotrophin-releasing hormone analogs can protect ovarian function, and gonadotrophin-releasing hormone analogs should be used with caution as an effective method to preserve fertility [6]. In addition, there are some novel strategies, such as stem cell therapy [6]. Mesenchymal stem cells (MSCs) are multipotent stem cells capable of cytokine secretion and immunomodulation. Stem cells have significant self-renewal capabilities and differentiate into specific tissues based on their surrounding environment and signals. Stem cells can regenerate and repair tissues by secreting cytokines and extracellular vesicles. They play an important therapeutic role in the fight against fibrosis in various organs[10]. Stem cells improve experimental colitis [11] and inhibit inflammation and fibrosis to prevent the progression of early diabetic nephropathy [12]. It has found that MSCs can reduce ROS accumulation and inhibit the AMP-dependent AMPK/mTOR signaling pathway to reduce the apoptosis of theca-interstitial cells and promote ovarian tissue repair [13]. Meanwhile, MSCs can also inhibit ovarian fibrosis through the AMPK/NR4A1 pathway [14]. However, stem cell therapy faces challenges, such as vascular embolism [15] and ethical concerns [16]. Therefore, studies have been conducted on extracellular vesicles (EVs). EVs are typically 30–200 nm in diameter and are released from multiple cell types. EVs are membrane-bound vesicles containing biomolecules such as microRNAs and proteins [17]. EV therapy can avoid many problems associated with stem cell therapy [18]. EVs have been widely used to treat multiple diseases, such as cerebral infarction [19], skin wound healing [20], and premature ovarian failure [21]. Some studies have found that EVs can upregulate the expression of Nrf2 in ovarian granulosa cells, promote the expression of oxidative stress-related proteins, inhibit cell apoptosis, and promote cell proliferation [22]. Other studies have found that human adipose-derived stem cells-EV can improve ovarian function, increase the number of follicles in premature ovarian insufficiency mice, and restore hormone levels to normal by regulating the SMAD signaling pathway [23]. However, conventional two-dimensional culture does not meet the requirements for optimal EV production. Three-dimensional (3D) culture can increase EV secretion and provide more efficacious EVs [24,25,26]. Therefore, we chose a 3D culture method to culture MSCs.
The ovaries have short blood flow, and the ovarian artery enters the ovarian hilus and branches within the ovary to supply both the cortex and medulla [27]. Thus, stem cells and EVs injected intravenously rarely reach the ovaries. Moreover, EVs injected intravenously, peritoneally, and subcutaneously are easily cleared by blood circulation [28], and EVs may accumulate in multiple non-target organs [29]. To solve these problems, some studies have used extracellular matrix (ECM) gels from the female reproduction system [30] or other gels [31, 32] to encapsulate EVs, concentrate EVs in the ovary, and sustain the release of EVs. However, no such studies have used a 3D-MSC-EVs-ECM gel to treat COF. Therefore, in the current study, we tested whether a 3D-MSC-EVs-ECM gel was effective in treating CTX-induced ovarian failure.
Materials and methods
Primary culture and identification of hUC-MSCs
The human umbilical cord (UC) was collected from healthy full-term fetuses that were born via cesarean delivery at Qilu Hospital of Shandong University. The use of the UC was approved by the Ethics Committee of the Qilu Hospital of Shandong University (KYLL-2021(KS)-086). Briefly, the UC was cut into small fragments (1–2 mm3) after the umbilical arteries and vein were dissected and placed in dishes with fresh MSC complete medium containing 90% α-MEM (Macgene Technology Ltd., Beijing, China), 10% fetal bovine serum (FBS; Bovogen Biologicals Pty Ltd, Keilor East, Victoria, Australia) and 1% penicillin and streptomycin (P/S; C125C5, New Cell & Molecular Biotech, Suzhou, China) in 5% CO2 at 37 ℃. When the cells reached 80%–90% confluence, they were trypsinized with 0.25% trypsin (C125C1, New Cell & Molecular Biotech) and passaged at a 1: 3 ratio. hUC-MSCs at passage three in the logarithmic growth period were collected to perform tri-lineage differentiation and phenotypic identification. The adipogenic, osteogenic, and chondrogenic abilities of the hUC-MSCs were detected after using the corresponding induction solution. After 4 weeks of induction, Oil Red O, Alizarin Red S, and Alcian Blue staining were used to detect adipogenic, osteogenic, and chondrogenic differentiation, respectively. In addition, hUC-MSCs were stained with antibodies against the human antigens CD29-PE (TS2/16, 303,004), CD31-PE (WM59, 303,106), CD44-PE (IM7, 25–0441-82), CD45-PE (2D1, 368,510), CD73-PE (AD2, 344,004), CD90-PE (eBio5E10,12–0909-42), CD105-PE (SN6, 12–1057-42), and CD271-PE (ME20.4, 12–9400-42). CD29, CD31, CD45, and CD73 antibodies were purchased from BioLegend (San Diego, CA, USA) and CD44, CD90, CD105, and CD271 antibodies were purchased from Invitrogen (Thermo Fisher Scientific, Inc.). The antibody dilution ratio was 1:20. The cells were then analyzed by flow cytometry (Guava easyCyte 6HT, EMD Millipore, Billerica, MA, USA), and the data were examined using FlowJo (version 10, Tree Star Inc., Ashland, OR, USA).
Three-dimensional culture of hUC-MSCs
hUC-MSCs (passages 3–6) were cultured in T75 cm2 flasks. For 3D culture, 5 × 106 hUC-MSCs (passages 3–6) were seeded in ultra-low attachment dishes (633,180, Greiner, Frickenhausen, Germany). 3D-cultured hUC-MSCs were stained with PKH26 (MIDI26, Sigma-Aldrich, USA) and 4′,6-diamidino-2-phenylindole (DAPI, ab104139, Abcam) and observed under a fluorescence microscope (BX53; Olympus, Tokyo, Japan).
Cell proliferation assay
Cell proliferation was determined using carboxyfluorescein diacetate succinimidyl ester for both 2D- and 3D-cultured cells (CFSE; BD Biosciences, San Jose, CA, USA). Cells were incubated for 5 min with 10 μM of CFSE in phosphate-buffered saline (PBS) at 37 ℃. All assays were performed using flow cytometry. Cell proliferation analyses were performed using ModFit LT software (version 3.2; Verity Software House, Topsham, ME, USA).
Quantitative real-time polymerase chain reaction (qRT-PCR) assay
Total RNA from 2D- and 3D-cultured cells was extracted using an RNA fast kit (Fastagen, Shanghai, China). Total RNA was reverse-transcribed using the ReverTra Ace qPCR RT Kit (Toyobo, Osaka, Japan). qRT-PCR was performed to detect the transcript levels of Oct4, Nanog, Igf, Sox2, Hgf, Tgfb1, Sdf, Vegf, Ido1, Il6 and Pge2 using an SYBR Green real-time PCR master mix (Toyobo, Osaka, Japan). Relative expression levels were normalized to those of GAPDH. The value for 2D-cultured cells was set to 1. Primer sequences used are listed in Supplementary Material 1.
Isolation and purification hUC-MSC-derived EVs
hUC-MSCs were cultured in flasks and ultra-low attachment dishes with EVs-depleted FBS. EVs-depleted FBS was prepared by separating EVs from FBS using ultracentrifugation at 100,000 × g for 18 h at 4 ℃ (Thermo Fisher Scientific, Sorvall Lynx 6000, Osterode, Germany) [33]. After 48 h, cell culture supernatants were collected to isolate the EVs. The 2D and 3D cell culture supernatants were centrifuged at 300 × g for 10 min, 2,000 × g for 20 min, and 10,000 × g for 30 min at 4 ℃ to eliminate the dead cells and cell debris. The supernatants were filtered using a 0.22-μm filter (Millipore). Then, the supernatants were centrifuged at 100,000 × g for 70 min, resuspended in PBS, and centrifuged again at 100,000 × g for 70 min at 4 ℃. Finally, EVs purified from the media of 2D- and 3D-cultured cells (2D-EVs and 3D-EVs, respectively) were resuspended in 100–200 μL of PBS and stored at -80 ℃.
Transmission electron microscopy (TEM)
The morphology of the EVs was assessed by TEM, which was conducted by Shanghai XP Biomed Co., Ltd. (Shanghai, China). Droplets were deposited onto a TEM copper grid for more than 1 min. The grid was then negatively stained with phosphotungstic acid for 5 min, blotted with filter paper, and dried at room temperature. Finally, the EVs were observed under a transmission electron microscope (JEM-1200EX, JEOL, Tokyo, Japan) and photographed.
Nanoparticle tracking analysis (NTA)
The size distribution and concentration of EVs were determined using NTA. The EV nanoparticle tracking analysis was conducted by Shanghai XP Biomed Co., Ltd. (Shanghai, China). Briefly, EVs were diluted in PBS and injected into the ZetaView particle tracker (ZetaView PMX 110, Particle Metrix, Meerbusch, Germany). NTA measurements were recorded and analyzed at 11 positions. ZetaView software (version 8.04.02 SP2) was used to analyze the data. We used the bicinchoninic acid assay (BCA) method to detect the protein concentration of EVs and combined it with NTA analysis to determine that there were 109 particles per μg of protein and 5 × 1011 particles per 0.5 mg of protein.
Preparation of ovarian ECM, lyophilized EVs, and gels
Five-week-old female Sprague–Dawley rats (Sibeifu Beijing Biotechnology Co. Ltd., Beijing, China) were euthanized for ovary collection. The ovaries were decellularized with 0.2% sodium dodecyl sulfate (SDS, Solarbio, Beijing, China) for 12 h at 18–20 ℃ on a horizontal shaker at 50 rpm. Then, the decellularized ovaries were washed in PBS eight times for 2 h. A Masson’s trichrome stain kit (G1340, Solarbio) was used to confirm that the ovaries were completely decellularized. The ovarian ECM was lyophilized using a Scientz − 12N (Ningbo, China). The lyophilized ovarian ECM was digested with pepsin (Solarbio) for 72 h at a final concentration of 10 mg/mL to obtain ovarian ECM solutions. A gel consisting of chitosan (419,419, Sigma-Aldrich, USA)/β-sodium glycerophosphate (Merck, Darmstadt, Germany)/gelatin (Amresco) loaded with ovarian ECM solutions was obtained as described by Xu et al. [31]. Then, 2D-EVs and 3D-EVs were lyophilized and mixed with the ECM gel at 0.5 mg/mL. The viscosity of the gel was measured using an NDJ-9S viscometer (Shanghai Performance Tai Electronic Technology Co., Ltd, Shanghai, China) at 60 rpm [34].
Isolation and characterization of rat ovarian granulosa cells (OGCs)
Rat OGCs were isolated from four-week-old female Sprague–Dawley rats as described previously [35]. The rat OGCs were cultured in DMEM/F12 (Macgene) medium with 10% FBS and 1% P/S at 37 ℃, 5% CO2. Immunofluorescence staining was used to identify the follicle-stimulating hormone receptor (FSHR, GB11275-1, Servicebio, Wuhan, China) specifically expressed in granulosa cells. The nuclei were stained with DAPI (ab104139, Abcam).
Labeling of EVs and cellular uptake in vitro
2D- and 3D-EVs were labeled with PKH26 (Sigma) according to the manufacturer’s instructions. These EVs were then co-cultured with OGCs for 24 h (the final concentration of EVs was 50 μg/mL). The nuclei of the OGCs were stained with DAPI (Abcam). The fluorescence signals of PKH26 and DAPI were observed under a fluorescence microscope.
Cell counting kit-8 assay (CCK-8) and cytocompatibility assay in vitro
Rat OGCs were cultured in 96-well plates to reach 70–80% confluence. Then, rat OGCs were divided into six groups: Control, ECM gel, CTX, CTX + ECM gel, CTX + 2D-MSC-EVs-ECM gel, and CTX + 3D-MSC-EVs-ECM gel groups (gel: media = 1:10; final concentration of CTX (PHR1404, Sigma-Aldrich, USA), 6 mg/mL; final concentration of EVs, 10 μg/well). After 48 h, cell viability was detected using a CCK-8 assay (C6005, New Cell & Molecular Biotech). Absorbance was measured with an enzyme-labeled analyzer (DNM-9602, Beijing Perlong New Technology Co., Ltd.) at a wavelength of 450 nm.
Establishment of COF rat model and therapeutic experiments
Five-week-old female Sprague–Dawley rats were obtained from Sibeifu Beijing Biotechnology Co., Ltd., Beijing, China. The use of rats was approved by the Ethics Committee of the Qilu Hospital of Shandong university (DWLL-2021–147). The rats were maintained at 22 ± 2 ℃ under a 12-h light/dark cycle, with free access to water and food. The rats were randomly divided into six groups (5–7 rats per group): Control, ECM gel, CTX, CTX + ECM gel, CTX + 2D-MSC-EVs-ECM gel, and CTX + 3D-MSC-EVs-ECM gel group. The Control group received no treatment. The first day of the CTX injection was designated as day 1. The CTX, CTX + ECM gel, CTX + 2D-MSC-EVs-ECM gel and CTX + 3D-MSC-EVs-ECM gel groups received intraperitoneal injections of CTX (50 mg/kg on day 1 and 8 mg/kg on days 2–14). The CTX group did not receive any treatment after 14 days. The ECM group received an intra-ovarian injection of ECM gel on day 15. The CTX + ECM gel, CTX + 2D-MSC-EVs-ECM gel, and CTX + 3D-MSC-EVs-ECM gel groups received intra-ovarian injections of ECM gel, 2D-MSC-EVs-ECM gel, and 3D-MSC-EVs-ECM gel, respectively, on day 15. Each ovary was injected with 25 μL of gel containing 0.5 mg/mL of EVs. All the rats were weighed daily. All vaginal smears were collected daily and stained with hematoxylin and eosin (H&E) to determine the estrous cycle phase. All rats were euthanized on day 30 for further analysis.
Enzyme-linked immunosorbent assay (ELISA)
Blood samples were obtained by cardiac puncture. Serum was collected by centrifuging at 1,200 × g for 15 min at 4 ℃, and it was stored at –20 ℃ for ELISA analysis. Serum levels of FSH (Cloud-clone Corp, CEA830Ra), estradiol (E2, Cloud-clone Corp, CEA461Ge), and anti-Mullerian hormone (AMH, Cloud-clone Corp, CEA228Ra) were detected using an ELISA kit according to the manufacturer’s instructions.
H&E staining
Rat ovaries were fixed in 4% paraformaldehyde, embedded in paraffin, and sectioned into 5-μm sections. Five discontinuous sections of each ovary were selected for follicle counting and morphological observation. The ovarian sections were dewaxed and rehydrated for H&E staining.
RNA sequencing
Ovary tissues (Control, CTX, CTX + 2D-MSC-EVs-ECM gel, and CTX + 3D-MSC-EVs-ECM gel groups) were chosen for RNA sequencing. Each group had one biological replicate. RNA sequencing and bioinformatics analyses were performed at Shandong Xiuyue Biotechnology Co. Ltd.
Western blotting
The protein lysates from exosomes or ovary tissues were prepared following standard protocols, and the protein content was determined using a BCA protein assay kit (P0010S, Beyotime, Shanghai, China). Equal amounts of proteins were electrophoresed on 7.5–15% SDS-PAGE gels and transferred onto polyvinylidene difluoride membranes (ISEQ00010, Merck Millipore, Darmstadt, Germany). Membranes were blocked for 2 h in blocking buffer composed of 5% non-fat dry milk in tris-buffered saline containing 0.1% Tween 20 at room temperature and were incubated with primary antibodies overnight at 4 ℃. The polyvinylidene difluoride membranes were then washed three times for 10 min each and incubated with secondary antibodies for 1 h at room temperature. A chemiluminescence detection system (Shanghai Jiapeng Technology Co., Ltd., Shanghai, China) was used to visualize the protein bands. Protein expression was expressed as relative abundance normalized to that of GAPDH (1:1000, 5174, Cell Signaling Technology). The primary antibodies used were anti-Alix (1:1000, 18269S, Cell Signaling Technology), anti-CD63 (1:1000, 25,682–1-AP, Proteintech), anti-CD9 (1:1000, 13174S, Cell Signaling Technology), anti-CD81 (1:1000, 52892S, Cell Signaling Technology), anti-TSG101 (1:1000, ab125011, Abcam, Cambridge, UK), anti-calnexin (1:1000, 2433S, Cell Signaling Technology), and anti-Bax (1:1000, 2772S, Cell Signaling Technology). The secondary antibody used was Goat Anti-Rabbit IgG (H + L) HRP (1:5000, AB0101, Abways).
Statistical analysis
All experiments were performed in triplicate. All data were analyzed using GraphPad Prism version 9 (GraphPad, San Diego, CA, USA). Comparisons between two groups or multiple groups were analyzed using Student’s t-test or one-way ANOVA. Quantitative data were expressed as mean ± standard deviation (SD). p < 0.05 was considered statistically significant.
Results
Morphology and identification of primary hUC-MSCs
After seven days, hUC-MSCs migrated out of the tissue and showed a typical fibroblast-like phenotype (Fig. 1A). Their ability to differentiate into adipocytes, osteoblasts, and chondrocytes was confirmed by specific staining. The lipid droplets of the adipocytes were stained red using Oil Red O, the calcified ECM of the osteoblasts was stained red using Alizarin Red S, and the proteoglycans of the chondrocytes were stained blue using Alcian Blue (Fig. 1B). Flow cytometry showed that isolated hUC-MSCs positively expressed CD29, CD44, CD73, CD90, and CD105 and negatively expressed CD31, CD45, and CD271 (Fig. 1C).
Identification of hUC-MSCs. A Microscopic image of hUC-MSCs. Scale bar: 200 μm. B hUC-MSCs differentiated into adipocytes, osteoblasts, or chondrocytes after corresponding induction, indicated by Oil Red O, Alizarin Red S, and Alcian Blue staining. Scale bars: 200 μm. C Detection of the specific markers of hUC-MSCs by flow cytometry illustrated that hUC-MSCs were positive for CD29, CD44, CD73, CD90, and CD105 and negative for CD31, CD45, and CD271
Morphology and function of hUC-MSCs in 3D culture
A schematic diagram of hUC-MSCs cultured in an ultra-low attachment dish is shown in Fig. 2A. After 48 h, hUC-MSCs aggregated into spheroids, the cell membranes were stained red with PKH26, and the nuclei were stained blue with DAPI (Fig. 2B). The diameters of the spheroids were approximately 200 μm. Compared with hUC-MSCs in 2D culture, hUC-MSCs in 3D culture had a significantly higher proliferation index (50.522 ± 2.843 vs. 38.279 ± 1.036, p < 0.0001) (Fig. 2C). Otherwise, the expression levels of nutritional cytokines and immunomodulatory and stemness-related genes were significantly upregulated in hUC-MSCs in 3D culture. These included Oct4, Nanog, Igf, Sox2, Hgf, Tgfb1, Sdf, Vegf, Ido1, Il6 and Pge2 (Fig. 2D).
Morphology, proliferation, and gene expression of 3D-cultured hUC-MSCs. A The schematic diagram of hUC-MSCs cultured in an ultra-low attachment dish. B hUC-MSCs in 3D culture. The diameter of the spheroid is approximately 200 μm. Scale bars: 50 μm. C Cell proliferation of hUC-MSCs in 2D and 3D culture (n = 3, ****p < 0.0001, Student’s t-test). D Expression of nutritional cytokine, immunomodulatory, and stemness-related genes (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, Student’s t-test)
Characterization of 2D-EVs and 3D-EVs
EVs were isolated by ultracentrifugation (Fig. 3A). 2D-EVs and 3D-EVs showed teacup-like double-sided structures (Fig. 3B). NTA analysis showed that the median diameters of the 2D-EVs and 3D-EVs were 125.9 nm and 119.3 nm, respectively (Fig. 3C). This was consistent with the general diameter criteria (30–200 nm). Western blotting results showed that exosomal markers Alix, TSG101, CD9, CD63, and CD81 were positively expressed in hUC-MSCs, 2D-EVs, and 3D-EVs. However, calnexin, which is expressed in hUC-MSCs, was negatively expressed in 2D-EVs and 3D-EVs (Fig. 3D). We compared the relationship between cell number and extracellular vesicle secretion. The concentration of 2D-EVs from 2 × 107 cells was 2.5 × 1010 particles/mL (250 particles/cell), and the concentration of 3D-EVs from 2 × 107 cells was 1.4 × 1011 particles/mL (1,400 particles/cell) (Fig. 3E). The concentration of EVs was measured using a BCA kit, and the 2D- and 3D-EVs were approximately 1.0 × 109 particles/μg. The lyophilized EVs are shown in Fig. 3F.
Characterization of EVs and gels. A EVs at the bottom of the ultracentrifuge tube. B Morphology of EVs under TEM. Scale bars: 100 μm. C Particle size distribution of EVs in NTA. D Identification of EVs by western blot. E The yield of EVs from 2D-EVs and 3D-EVs. F Appearance of lyophilized EVs. G Decellularization of ovaries. H Ovarian ECM and control ovary were stained with Masson’s trichrome stain kit. Ovarian ECM was only left with collagen which was stained blue. Scale bars: 100 μm. I Appearance of lyophilized ovarian ECM. J ovarian ECM digestion. K ECM gel. L ECM gel after 37 ℃
Preparation and characterization of ovarian ECM gel
The ovaries were decellularized using 0.2% SDS (Fig. 3G). Masson’s trichrome staining revealed only collagen fibers in the decellularized ovary, which stained blue (Fig. 3H). However, collagen fibers, muscle fibers, and nuclei were stained and observed in the control ovary, which was stained blue, red, and blue-black (Fig. 3H). This demonstrated that the ovaries were completely decellularized. The lyophilized and digested ovarian ECM are shown in Fig. 3I and J. Gel solutions were prepared using chitosan/β-sodium glycerophosphate/gelatin loaded with ovarian ECM solutions (Fig. 3K). The gel solutions formed gel in 37 ℃ (Fig. 3L). The viscosity of the gel was 98 mPa·s.
Identification of rat OGCs and enhanced therapeutic efficacy of 3D-MSC-EVs-ECM gel in CTX-treated OGCs
Rat ovarian granulosa cells were extracted from the rat ovaries. The morphology of the rat OGCs was fibroblastic (Fig. 4A). Immunofluorescence staining revealed that FSHR was specifically expressed in the cytoplasm of the OGCs (Fig. 4B). The cytoplasm was stained green, and the nuclei were stained blue. Moreover, 2D-EVs and 3D-EVs were successfully taken up by the OGCs (Fig. 4C). The nuclei of the OGCs were stained blue. The CCK-8 assay showed that the ECM gel did not affect normal rat OGCs, indicating that the ECM gel had good cytocompatibility. The viability of the CTX-treated OGCs was significantly enhanced by treatment with the 2D-MSC-EVs-ECM and 3D-MSC-EVs-ECM gels compared with the ECM gel, and the 3D-MSC-EVs-ECM gel was more effective (Fig. 4D).
Identification of rat OGCs and enhanced efficacy of treatment with the 3D-MSC-EVs-ECM gel. A Morphology of rat OGCs. Scale bars: 200 μm. B Identification of rat OGCs. Rat OGCs positively expressed FSHR. Scale bars: 200 μm. C EVs were effectively taken up by rat OGCs. Scale bars: 200 μm. D CCK-8 assay in rat OGCs (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001, Student’s t-test)
Enhanced therapeutic efficacy of 3D-MSC-EVs-ECM gel in CTX-treated rat model
The procedures for animal experiments are described in Fig. 5A. Intraovarian injection of 25 μL of gel did not cause obvious tissue swelling or bleeding of the ovaries (Fig. 5B). At the beginning of the experiment, all rats in each group had similiar body weights; however, their body weights changed at day 30 (Fig. 5C). The Control and ECM gel groups had similar body weights, whereas the body weights of the other groups decreased. The CTX group showed the greatest decrease. The body weight of the CTX + ECM gel group recovered to a greater extent than that of the CTX group. The body weights of the CTX + 2D-MSC-EVs-ECM gel group and CTX + 3D-MSC-EVs-ECM gel group recovered more than those of the CTX + ECM gel group. However, the CTX + 3D-MSC-EVs-ECM gel group showed the best recovery. The estrus cycles of the rats were monitored, and they had regular estrus cycles at the beginning of the experiments (Fig. 5D). The CTX group had a longer estrous cycle for days after CTX injury. However, the CTX + 2D-MSC-EVs-ECM gel group and the CTX + 3D-MSC-EVs-ECM gel group recovered (Fig. 5E).
Animal experimental procedures and the weights and estrous cycle days in the rat model. A Schematic illustration of the procedures of animal experiments. B Intra-ovarian injection and after injection. C Body weight in each group before rats were sacrificed (n = 5–7; *p < 0.05, **p < 0.01, Student’s t-test). D Regular estrous cycles. a: Proestrus; b: Estrus; c: Metestrus; d: Diestrus. Scale bars: 200 μm (E) Estrous cycles in each group (n = 5–7; *p < 0.05, **p < 0.01, Student’s t-test)
For primordial, primary, secondary, and preovulatory follicles, there were no differences between the Control and the ECM gel groups. There were a minimum number of primordial and secondary follicles in the CTX group. Furthermore, compared to the CTX group, the numbers of primordial follicles and secondary follicles had increased in the CTX + ECM gel, CTX + 2D-MSC-EVs-ECM gel, and CTX + 3D-MSC-EVs-ECM gel groups. The CTX + 3D-MSC-EVs-ECM gel group had the most primordial and secondary follicles. However, there were no differences in the number of primary follicles between the CTX + 2D-MSC-EVs-ECM gel and CTX + 3D-MSC-EVs-ECM gel groups (Fig. 6A).
H&E staining of ovaries, ELISA, and expression of Bax. A Representative images and analysis of H&E staining in the ovaries of each group. Scale bars: 200 μm. n = 3; *p < 0.05, **p < 0.01, ***p < 0.001, Student’s t-test. B, C, D The serum levels of AMH, E2, and FSH in each group (n = 3; ***p < 0.001, ****p < 0.0001, Student’s t-test). E Western blotting analysis of Bax in each group (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001, Student’s t-test)
The hormones indicative of ovarian function, AMH and E2, decreased, and FSH levels increased significantly with CTX treatment. In comparison, treatment with the CTX + 2D-MSC-EVs-ECM gel and the CTX + 3D-MSC-EVs-ECM gel gave less of a reduction in AMH and E2 and less of an increase in FSH than treatment with CTX alone. In particular, the CTX + 3D-MSC-EVs-ECM gel group recovered significantly (Fig. 6B–D).
Bax expression was detected by western blot analysis. There was no difference between the Control and ECM gel groups. The expression of Bax was the highest in the CTX group. The expression of Bax decreased in the CTX + ECM gel, CTX + 2D-MSC-EVs-ECM gel, and CTX + 3D-MSC-EVs-ECM gel groups compared to that in the CTX group (Fig. 6E).
RNA sequencing of rat ovaries
We named the Control group [Con], the CTX group [CTX], the CTX + 2D-MSC-EVs-ECM gel group [2D], the CTX + 3D-MSC-EVs-ECM gel group [3D]. Venn diagram software was used to analyze the upregulated and downregulated differentially expressed genes in CTX vs. Con and 2D vs. CTX (Fig. 7A) to detect the influence of the 2D-MSC-EVs-ECM gel on the CTX-treated ovaries, CTX vs. Con and 3D vs. CTX (Fig. 7B) to detect the influence of the 3D-MSC-EVs-ECM gel on the CTX-treated ovaries, and 2D vs. CTX and 3D vs. CTX to detect the difference in the effect between 2D-MSC-EVs-ECM gel and 3D-MSC-EVs-ECM gel (Supplementary Material 2) (Fig. 8C). Moreover, we compared the top 20 upregulated and downregulated genes in CTX vs. Con, 2D vs. CTX, and 3D vs. CTX, with counts ≥ 100 as a standard. We found that Hbb-b1 was upregulated in CTX vs. Con and downregulated in 2D vs. CTX and 3D vs. CTX; Gpd1 was upregulated in CTX vs. Con and downregulated in 3D vs. CTX; ENSRNOG00000013279 and Testin were upregulated in CTX vs. Con and downregulated in 2D vs. CTX; and Clec4a3 and Sirpa were downregulated in CTX vs. Con and upregulated in 3D vs. CTX. We also found that ENSRNOG00000062337, Irf7, Oasl2, Cxcl10, Lyz2, Mpeg1, Rt1-s3, and Lilrb4 were upregulated in 2D vs. CTX and 3D vs. CTX, and Col7al, Ptprn, and Nap1l5 were downregulated in 2D vs. CTX and 3D vs. CTX. Furthermore, we screened for differentially expressed genes between the CTX + 3D-MSC-EVs-ECM gel group and the CTX + 2D-MSC-EVs-ECM gel group with a criterion of |log2 (fold change)|≥ 1, p < 0.05, and counts ≥ 100. There were 233 differentially expressed genes, including 89 upregulated and 144 downregulated genes (Supplementary Material 3). The Kyoto Encyclopedia of Genes and Genomes pathway enrichment results showed that the top three pathways enriched for differentially expressed genes were cytokine-cytokine receptor interaction (map04060), neuroactive ligand-receptor interaction (map04080), and human papillomavirus infection (map05165) (|log2 (fold change)|≥ 1, p < 0.05) (Fig. 8A). The Gene Ontology (GO) analysis revealed that the top three pathways were enriched for developmental process (GO:0032502), membrane part (GO:0044425), and protein binding (GO:0005515) (|log2 (Fold change)|≥ 1, p < 0.05) (Fig. 8B–D).
Discussion
Chemotherapy-induced ovarian failure is a long-term adverse effect of chemotherapy drugs, especially alkylating agents [9].It disrupts endocrine and reproductive ovarian function and has become increasingly common in recent years. There are many methods to prevent and treat COF [6]; however, these methods have shortcomings. Stem cells have shown promise. However, owing to the ethical and safety issues of stem cells, EVs derived from mesenchymal stem cells have been used as an alternative therapy in regeneration medicine [36] and play important roles in many diseases. In our study, we used ultra-low attachment dishes to culture hUC-MSCs and produced 2D- and 3D-EVs. We then mixed these EVs with chitosan/β-sodium glycerophosphate/gelatin loaded with ovarian ECM solutions to obtain 2D- and 3D-MSC-EVs-ECM gels. We found that the 3D-MSC-EVs-ECM gel most significantly restored the cell viability of CTX-treated rat OGCs. In CTX-treated rats, the 3D-MSC-EVs-ECM gel increased the serum levels of AMH and E2 and decreased the serum levels of FSH. According to H&E staining, the 3D-MSC-EVs-ECM gel promoted an increase in the number of primordial follicles. The 3D-MSC-EVs-ECM gel decreased the expression levels of the apoptosis-related gene Bax, according to western blot analysis.
To increase the output of EVs, we used low attachment dishes to culture the hUC-MSCs. Methods for 3D culture are classified as scaffold-free or scaffold-based [37]. 3D culture can simulate the in vivo microenvironment [38]. 3D culture refers to a culture method that uses materials such as scaffolds and collagen to provide cells with a microenvironment that is closer to the living conditions in the body and can increase the communication between cells and between cells and the extracellular matrix. 3D culture can simulate the biochemical and biomechanical environment in the body, make the cytoskeleton more consistent with the reality of cells in vivo, promote the self-renewal of stem cells and inhibit their differentiation [39, 40]. 3D culture enables cells to grow and migrate in all three-dimensional spaces, which is beneficial for the molecular transfer and material exchange between cells and between cells and the extracellular matrix, and is beneficial for promoting the secretion of exosomes by cells. Ultra-low attachment dishes are scaffold free. After 48 h of 3D culture, hUC-MSCs aggregated into spheroids, and the proliferation index of the hUC-MSCs increased compared to that in 2D culture. Surprisingly, nutritional cytokines, immunomodulatory, and stemness-related genes were significantly upregulated in hUC-MSCs after 3D culture. This reflected that the immunomodulatory function, antiapoptotic function, and angiogenesis of hUC-MSCs in 3D culture were upregulated. It has been demonstrated that exosomes from 3D-cultured human embryonic stem cell can improve liver fibrosis by targeting the TGFβRII-SMADS pathway [41] and improve memory and cognitive deficits in mice with Alzheimer’s disease [42]. For EVs, the particles/cell in 3D culture were 5.6 times more than in 2D culture. The yield of EVs increased after 3D culture, which was consistent with previous studies [25, 26]. The ovary is a reproductive organ, and it has better safety than DNA-free EVs in treatment to avoid foreign DNA contamination.
Recently, it was shown that bone marrow MSC-derived exosomes can inhibit the apoptosis of cisplatin-induced ovarian granulosa cells [43], recover the estrus cycle, and increase the number of basal follicles in premature ovarian failure [35]. However, the use of EVs has many risks, as previously mentioned. Hydrogels can prolong the release of exosomes and increase their retention in vivo [32]. Organ-specific ECMs have been widely used in recent years. The ECM is a non-cellular 3D network that consists of collagen, elastin, and laminin and provides structural and functional support [44]. ECMs have emerging applications in gynecology, such as uterine ECM [45], ovarian ECM [46], oviduct ECM [47], and vagina ECM [48]. In our study, we mixed chitosan/β-sodium glycerophosphate/gelatin with ovarian ECM solutions to prepare a gel. Chitosan has been widely used for gel preparation [49]. Gelatin was used to minimize gelation time. Finally, we demonstrated that the gel itself was not harmful to rats, and the 3D-MSC-EVs-ECM gel improved multiple endpoints in CTX-treated rats.
We used ovarian tissue (Control, CTX, CTX + 2D-MSC-EVs-ECM gel, CTX + 3D-MSC-EVs-ECM gel groups) for RNA sequencing. As shown in Fig. 7A, 502 genes were differentially expressed between CTX_Con_up and 2D_CTX_down, and 1021 genes were differentially expressed between CTX_Con_down and 2D_CTX_up. As shown in Fig. 7B, 622 genes were differentially expressed between CTX_Con_up and 3D_CTX_down, and 1017 genes were differentially expressed between CTX_Con_down and 3D_CTX_up. As shown in Fig. 7C, 973 genes were differentially expressed between 2 and 3D_CTX_up, and 630 genes were differentially expressed between 2 and 3D_CTX_down. These overlapping genes need to be further analyzed and studied.
In our study, we found that the expression of Hbb-b1 increased in CTX-treated rats but decreased in CTX + 2D-MSC-EVs-ECM gel and CTX + 3D-MSC-EVs-ECM gel groups. It has been demonstrated that capsaicin can stimulate the expression of erythroid-specific genes, such as Hbb-b1, to induce the development of the erythroid lineage from bone marrow cells [50]. Hbb-b1 was downregulated in mice with intratracheal instillation of silver nanoparticles (AgNPs) because AgNPs may induce Th2-type dominant inflammatory responses and tissue damage in the lungs of mice [51]. In addition, chronic stress can affect the expression of genes involved in vascular system function, such as Hbb-b1, Hba-a2 [52], and Hbb-b1, and these can be potential markers of chronic social stress in mice [53]. Hba-a2 expression was also downregulated in the CTX + 3D-MSC-EVs-ECM gel group as compared to that in CTX treatment alone. Thus, we speculated that Hbb-b1 and Hba-a2 might be related to changes in ovarian vascular stress. Testin, a Sertoli cell secretory protein, was upregulated in the CTX group and downregulated in the CTX + 2D-MSC-EVs-ECM gel group. Some studies had shown that testin is involved in the epithelial-mesenchymal transition of endometrial cancer [54].
Gpd1 is a key enzyme regulating glycolysis and glycerol metabolism, and Gpd1 was upregulated in the CTX group and downregulated in the CTX + 3D-MSC-EVs-ECM gel group. Gpd1 may play an important role in hypoxia and lipid metabolism pathways in clear cell renal cell carcinoma [55], and Gpd1 may be a novel tumor suppressor in bladder cancer [56]. Sirpa was downregulated in the CTX group and upregulated in the CTX + 3D-MSC-EVs-ECM gel group. The CD47-SIRPA axis can mediate cancer cell immune escape and immunotherapy [57] and inhibit inside-out activation of integrin signaling in macrophages to suppress phagocytosis [58]. According to the enriched pathways of differentially expressed genes between the CTX + 3D-MSC-EVs-ECM gel group and CTX + 2D-MSC-EVs-ECM gel group, a cytokine-cytokine receptor interaction is involved in the estrous cycle in the ovaries at proestrus and estrus [59], and the neuroactive ligand-receptor interaction pathway is enriched in the differentially expressed genes between the low and the high egg production groups [60]. The differentially expressed genes could help us to identify novel targets and mechanisms for COF and how EVs function. This merits future research.
In summary, we have developed a new approach to use EVs to treat multiple diseases. Ovarian-related diseases could be treated by the injection of a 3D-MSC-EVs-ECM gel in the posterior fornix under the guidance of B-ultrasound in the future. However, several aspects of this approach could benefit from further research. Novel 3D culture methods need to be developed to improve the yield and performance of the EVs derived from them. In addition to gels, new carries need to be advanced to improve the release profile and preserve the functionality of EVs.
Conclusions
In our study, the output of EVs derived from mesenchymal stem cells increased in 3D culture compared with that in conventional 2D culture. The expression of nutritional cytokines and immunomodulatory and stemness-related genes were significantly upregulated in hUC-MSCs after 3D culture. We demonstrated that a 3D-MSC-EVs-ECM gel could recover the cell viability of CTX-treated OGCs in vitro and improve ovarian function by regulating the serum levels of AMH, E2, and FSH and the number of follicles in CTX-treated rats. Our study provides a novel cell-free therapy to treat COF and warrants further research on other diseases. Also, our study provides new evidence for the widespread development of 3D stem cell culture in the future. We chose hydrogel to encapsulate EVs, which is beneficial to protect EVs from rapid clearance, and can use the 3D grid structure and degradation process of hydrogel to regulate the sustained release of EVs, providing a new way for the targeted application of EVs. The differential genes and pathways we discovered through sequencing are helpful to find the differences between EVs derived from 3 and 2D cultured UC-MSCs, providing new targets and treatments for the treatment of COF. The in-situ injection method also provides a new way to treat ovarian-related diseases in the future, for example, injecting EV and ECM into the uterine cavity can be used to treat thin endometrium. This method can be performed under B-ultrasound guidance to avoid damage caused by surgery.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- COF:
-
Chemotherapy-induced ovarian failure
- CTX:
-
Cyclophosphamide
- EVs:
-
Extracellular vesicles
- hUC-MSCs:
-
Human umbilical cord mesenchymal stem cells
- MSCs:
-
Mesenchymal stem cells
- 3D:
-
Three-dimensional
- 2D:
-
Two-dimensional
- UC:
-
Umbilical cord
- ECM:
-
Extracellular matrix
- FBS:
-
Fetal bovine serum
- P/S:
-
Penicillin and streptomycin
- OGCs:
-
Ovarian granulosa cells
- DAPI:
-
4′,6-diamidino-2-phenylindole
- CFSE:
-
Carboxyfluorescein diacetate succinimidyl ester
- PBS:
-
Phosphate-buffered saline
- qRT-PCR:
-
Quantitative real-time polymerase chain reaction
- TEM:
-
Transmission electron microscopy
- NTA:
-
Nanoparticle tracking analysis
- BCA:
-
Bicinchoninic acid assay
- SDS:
-
Sodium dodecyl sulfate
- FSHR:
-
Follicle-stimulating hormone receptor
- H&E:
-
Hematoxylin and eosin
- ELISA:
-
Enzyme-linked immunosorbent assay
- CCK-8:
-
Cell counting kit-8 assay
- AMH:
-
Anti-Mullerian hormone
- E2:
-
Estradiol
- FSH:
-
Follicle-stimulating hormone
- α-MEM:
-
α-Minimal essential medium
- DMEM/F12:
-
Dulbecco's modified eagle medium/nutrient mixture F-12
- Gapdh:
-
Glyceraldehyde-3-phosphate dehydrogenase
- Alix:
-
Apoptotic linked-gene-product 2 (ALG-2) interacting protein X
- TSG101:
-
Tumor susceptibility gene 101
- Bax:
-
BCL2 associated X
- KEGG:
-
Kyoto encyclopedia of genes and genomes
- GO:
-
Gene ontology
References
Ni X, Li Z, Li X, Zhang X, Bai G, Liu Y, et al. Socioeconomic inequalities in cancer incidence and access to health services among children and adolescents in China: a cross-sectional study. Lancet. 2022;400(10357):1020–32.
Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33.
Yum S, Li M, Chen ZJ. Old dogs, new trick: classic cancer therapies activate cGAS. Cell Res. 2020;30(8):639–48.
Smith RA, Andrews KS, Brooks D, Fedewa SA, Manassaram-Baptiste D, Saslow D, et al. Cancer screening in the United States, 2017: a review of current American Cancer Society guidelines and current issues in cancer screening. CA Cancer J Clin. 2017;67(2):100–21.
van Dorp W, Haupt R, Anderson RA, Mulder RL, van den Heuvel-Eibrink MM, van Dulmen-den BE, et al. Reproductive function and outcomes in female survivors of childhood, adolescent, and young adult cancer: a review. J Clin Oncol. 2018;36(21):2169–80.
Xiong J, Xue L, Li Y, Tang W, Chen D, Zhang J, et al. Therapy of endocrine disease: Novel protection and treatment strategies for chemotherapy-associated ovarian damage. Eur J Endocrinol. 2021;184(5):R177–92.
Spears N, Lopes F, Stefansdottir A, Rossi V, De Felici M, Anderson RA, et al. Ovarian damage from chemotherapy and current approaches to its protection. Hum Reprod Update. 2019;25(6):673–93.
Mauri D, Gazouli I, Zarkavelis G, Papadaki A, Mavroeidis L, Gkoura S, et al. Chemotherapy associated ovarian failure. Front Endocrinol (Lausanne). 2020;11:572388.
Cui W, Stern C, Hickey M, Goldblatt F, Anazodo A, Stevenson WS, et al. Preventing ovarian failure associated with chemotherapy. Med J Aust. 2018;209(9):412–6.
Guo Y, Xue L, Tang W, Xiong J, Chen D, Dai Y, et al. Ovarian microenvironment: challenges and opportunities in protecting against chemotherapy-associated ovarian damage. Human Reproduction Update. 2024:30(5):614–47.
Yang F, Ni B, Liu Q, He F, Li L, Zhong X, et al. Human umbilical cord-derived mesenchymal stem cells ameliorate experimental colitis by normalizing the gut microbiota. Stem Cell Res Ther. 2022;13(1):475.
Xiang E, Han B, Zhang Q, Rao W, Wang Z, Chang C, et al. Human umbilical cord-derived mesenchymal stem cells prevent the progression of early diabetic nephropathy through inhibiting inflammation and fibrosis. Stem Cell Res Ther. 2020;11(1):336.
Lu X, Bao H, Cui L, Zhu W, Zhang L, Xu Z, et al. hUMSC transplantation restores ovarian function in POI rats by inhibiting autophagy of theca-interstitial cells via the AMPK/mTOR signaling pathway. Stem Cell Res Ther. 2020;11(1):268.
Cui L, Bao H, Zhu W, Tang Y, Luo Q, Si Y, et al. hUMSCs transplantation regulates AMPK/NR4A1 signaling axis to inhibit ovarian fibrosis in POI rats. Stem Cell Rev Rep. 2023;19(5):1449–65.
Cui LL, Kerkela E, Bakreen A, Nitzsche F, Andrzejewska A, Nowakowski A, et al. The cerebral embolism evoked by intra-arterial delivery of allogeneic bone marrow mesenchymal stem cells in rats is related to cell dose and infusion velocity. Stem Cell Res Ther. 2015;6:11.
Volarevic V, Markovic BS, Gazdic M, Volarevic A, Jovicic N, Arsenijevic N, et al. Ethical and Safety issues of stem cell-based therapy. Int J Med Sci. 2018;15(1):36–45.
Gyorgy B, Szabo TG, Pasztoi M, Pal Z, Misjak P, Aradi B, et al. Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles. Cell Mol Life Sci. 2011;68(16):2667–88.
Nikfarjam S, Rezaie J, Zolbanin NM, Jafari R. Mesenchymal stem cell derived-exosomes: a modern approach in translational medicine. J Transl Med. 2020;18(1):449.
Li H, Luo Y, Liu P, Liu P, Hua W, Zhang Y, et al. Exosomes containing miR-451a is involved in the protective effect of cerebral ischemic preconditioning against cerebral ischemia and reperfusion injury. CNS Neurosci Ther. 2021;27(5):564–76.
Bian D, Wu Y, Song G, Azizi R, Zamani A. The application of mesenchymal stromal cells (MSCs) and their derivative exosome in skin wound healing: a comprehensive review. Stem Cell Res Ther. 2022;13(1):24.
Liu M, Qiu Y, Xue Z, Wu R, Li J, Niu X, et al. Small extracellular vesicles derived from embryonic stem cells restore ovarian function of premature ovarian failure through PI3K/AKT signaling pathway. Stem Cell Res Ther. 2020;11(1):3.
Zhang L, Ma Y, Xie X, Du C, Zhang Y, Qin S, et al. Human pluripotent stem cell-mesenchymal stem cell-derived exosomes promote ovarian granulosa cell proliferation and attenuate cell apoptosis induced by cyclophosphamide in a POI-like mouse model. Molecules. 2023;28(5):2112.
Lin J, Wang Z, Huang J, Tang S, Saiding Q, Zhu Q, et al. Microenvironment-protected exosome-hydrogel for facilitating endometrial regeneration, fertility restoration, and live birth of offspring. Small. 2021;17(11):e2007235.
Mo M, Zhou Y, Li S, Wu Y. Three-dimensional culture reduces cell size by increasing vesicle excretion. Stem Cells. 2018;36(2):286–92.
Cao J, Wang B, Tang T, Lv L, Ding Z, Li Z, et al. Three-dimensional culture of MSCs produces exosomes with improved yield and enhanced therapeutic efficacy for cisplatin-induced acute kidney injury. Stem Cell Res Ther. 2020;11(1):206.
Haraszti RA, Miller R, Stoppato M, Sere YY, Coles A, Didiot MC, et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Mol Ther. 2018;26(12):2838–47.
Hossain MI, O’Shea JD. The vascular anatomy of the ovary and the relative contribution of the ovarian and uterine arteries to the blood supply of the ovary in the guinea-pig. J Anat. 1983;137(Pt 3):457–66.
Takahashi Y, Nishikawa M, Shinotsuka H, Matsui Y, Ohara S, Imai T, et al. Visualization and in vivo tracking of the exosomes of murine melanoma B16-BL6 cells in mice after intravenous injection. J Biotechnol. 2013;165(2):77–84.
Smyth T, Kullberg M, Malik N, Smith-Jones P, Graner MW, Anchordoquy TJ. Biodistribution and delivery efficiency of unmodified tumor-derived exosomes. J Control Release. 2015;199:145–55.
Frances-Herrero E, Rodriguez-Eguren A, Gomez-Alvarez M, de Miguel-Gomez L, Ferrero H, Cervello I. Future challenges and opportunities of extracellular matrix hydrogels in female reproductive medicine. Int J Mol Sci. 2022;23(7):3765.
Xu X, Gu Z, Chen X, Shi C, Liu C, Liu M, et al. An injectable and thermosensitive hydrogel: Promoting periodontal regeneration by controlled-release of aspirin and erythropoietin. Acta Biomater. 2019;86:235–46.
Zhao X, Liu Y, Jia P, Cheng H, Wang C, Chen S, et al. Chitosan hydrogel-loaded MSC-derived extracellular vesicles promote skin rejuvenation by ameliorating the senescence of dermal fibroblasts. Stem Cell Res Ther. 2021;12(1):196.
Tian Y, Gong M, Hu Y, Liu H, Zhang W, Zhang M, et al. Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry. J Extracell Vesicles. 2020;9(1):1697028.
Zhou P, Li X, Zhang B, Shi Q, Li D, Ju X. A human umbilical cord mesenchymal stem cell-conditioned medium/chitosan/collagen/beta-glycerophosphate thermosensitive hydrogel promotes burn injury healing in mice. Biomed Res Int. 2019;2019:5768285.
Yang M, Lin L, Sha C, Li T, Zhao D, Wei H, et al. Bone marrow mesenchymal stem cell-derived exosomal miR-144-5p improves rat ovarian function after chemotherapy-induced ovarian failure by targeting PTEN. Lab Invest. 2020;100(3):342–52.
Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther. 2018;9(1):63.
Duval K, Grover H, Han LH, Mou Y, Pegoraro AF, Fredberg J, et al. Modeling physiological events in 2D vs. 3D cell culture. Physiology (Bethesda). 2017;32(4):266–77.
Kim W, Gwon Y, Park S, Kim H, Kim J. Therapeutic strategies of three-dimensional stem cell spheroids and organoids for tissue repair and regeneration. Bioact Mater. 2023;19:50–74.
Wu X, Su J, Wei J, Jiang N, Ge X. Recent advances in three-dimensional stem cell culture systems and applications. Stem Cells Int. 2021;2021:9477332.
Duval K, Grover H, Han LH, Mou Y, Pegoraro AF, Fredberg J, et al. Modeling physiological events in 2D vs. 3D cell culture. Physiology (Bethesda, Md). 2017;32(4):266–77.
Wang N, Li X, Zhong Z, Qiu Y, Liu S, Wu H, et al. 3D hESC exosomes enriched with miR-6766-3p ameliorates liver fibrosis by attenuating activated stellate cells through targeting the TGFbetaRII-SMADS pathway. J Nanobiotechnol. 2021;19(1):437.
Yang L, Zhai Y, Hao Y, Zhu Z, Cheng G. The regulatory functionality of exosomes derived from hUMSCs in 3D culture for alzheimer’s disease therapy. Small. 2020;16(3):e1906273.
Sun B, Ma Y, Wang F, Hu L, Sun Y. miR-644-5p carried by bone mesenchymal stem cell-derived exosomes targets regulation of p53 to inhibit ovarian granulosa cell apoptosis. Stem Cell Res Ther. 2019;10(1):360.
Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. Extracellular matrix structure. Adv Drug Deliv Rev. 2016;97:4–27.
Lopez-Martinez S, Rodriguez-Eguren A, de Miguel-Gomez L, Frances-Herrero E, Faus A, Diaz A, et al. Bioengineered endometrial hydrogels with growth factors promote tissue regeneration and restore fertility in murine models. Acta Biomater. 2021;135:113–25.
Chiti MC, Vanacker J, Ouni E, Tatic N, Viswanath A, des Rieux A, et al. Ovarian extracellular matrix-based hydrogel for human ovarian follicle survival in vivo: a pilot work. J Biomed Mater Res B Appl Biomater. 2022;110(5):1012–22.
Frances-Herrero E, De Miguel-Gomez L, Lopez-Martinez S, Campo H, Garcia-Dominguez X, Diretto G, et al. Development of decellularized oviductal hydrogels as a support for rabbit embryo culture. Reprod Sci. 2021;28(6):1644–58.
Hou C, Zheng J, Li Z, Qi X, Tian Y, Zhang M, et al. Printing 3D vagina tissue analogues with vagina decellularized extracellular matrix bioink. Int J Biol Macromol. 2021;180:177–86.
Mei E, Chen C, Li C, Ding X, Chen J, Xi Q, et al. Injectable and biodegradable chitosan hydrogel-based drug depot contributes to synergistic treatment of tumors. Biomacromol. 2021;22(12):5339–48.
Lee SA, Ryu YS, Choi HI, Han IS. Capsaicin promotes the development of burst-forming units–erythroid (BFU-E) from mouse bone marrow cells. Exp Mol Med. 2007;39(3):278–83.
Park EJ, Choi K, Park K. Induction of inflammatory responses and gene expression by intratracheal instillation of silver nanoparticles in mice. Arch Pharm Res. 2011;34(2):299–307.
Stankiewicz AM, Goscik J, Majewska A, Swiergiel AH, Juszczak GR. The effect of acute and chronic social stress on the hippocampal transcriptome in mice. PLoS ONE. 2015;10(11):e0142195.
Stankiewicz AM, Goscik J, Swiergiel AH, Majewska A, Wieczorek M, Juszczak GR, et al. Social stress increases expression of hemoglobin genes in mouse prefrontal cortex. BMC Neurosci. 2014;15:130.
Dong R, Pu H, Wang Y, Yu J, Lian K, Mao C. TESTIN was commonly hypermethylated and involved in the epithelial-mesenchymal transition of endometrial cancer. APMIS. 2015;123(5):394–400.
Liu R, Feng Y, Deng Y, Zou Z, Ye J, Cai Z, et al. A HIF1alpha-GPD1 feedforward loop inhibits the progression of renal clear cell carcinoma via mitochondrial function and lipid metabolism. J Exp Clin Cancer Res. 2021;40(1):188.
Zhang W, He X, Yin H, Cao W, Lin T, Chen W, et al. Allosteric activation of the metabolic enzyme GPD1 inhibits bladder cancer growth via the lysoPC-PAFR-TRPV2 axis. J Hematol Oncol. 2022;15(1):93.
Jia X, Yan B, Tian X, Liu Q, Jin J, Shi J, et al. CD47/SIRPalpha pathway mediates cancer immune escape and immunotherapy. Int J Biol Sci. 2021;17(13):3281–7.
Morrissey MA, Kern N, Vale RD. CD47 ligation repositions the inhibitory receptor SIRPA to suppress integrin activation and phagocytosis. Immunity. 2020;53(2):290-302 e6.
Yang S, Zhou X, Pei Y, Wang H, He K, Zhao A. Identification of differentially expressed genes in porcine ovaries at proestrus and estrus stages using RNA-Seq technique. Biomed Res Int. 2018;2018:9150723.
Mu R, Yu YY, Gegen T, Wen D, Wang F, Chen Z, et al. Transcriptome analysis of ovary tissues from low- and high-yielding Changshun green-shell laying hens. BMC Genomics. 2021;22(1):349.
Acknowledgements
The authors sincerely thank Mr. Shizhe Liu and Mr. Peifeng Ji for their guidance in the analysis of gene sequencing data.
Funding
Natural Science Foundation of Shandong Province, No. ZR2023MH079; JiNan Science and Technology Bureau, No. JNKCJN202201; Natural Science Foundation of Shandong Province, No. ZR2020MH063; Shandong Province Enterprise Innovation Ability Enhancement Project, No. 2022TSGC2001.
Author information
Authors and Affiliations
Contributions
Yaping Zhang and Dong Li: Investigation, Methodology, Visualization, Writing - Original Draft. Yi Han, Min Wu and Shule Zhang: Investigation, Methodology. Huixian Ma and Linghong Liu: Validation, Project administration, Conceptualization, Supervision, Writing - Review & Editing. Xiuli Ju: Conceptualization, Funding acquisition, Writing - Review & Editing.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
The use of the umbilical cords was approved by the Ethics Committee of the Qilu Hospital of Shandong University (Title: Study on the treatment of chronic inflammatory diseases by mesenchymal stem cells; Approval document of Ethics Committee: Qilu Hospital of Shandong university; IACUC Issue No.: KYLL-2021(KS)-086; Approval date: February 20, 2021). The use of rats was approved by the Ethics Committee of the Qilu Hospital of Shandong University (Title: Study on the treatment of premature ovarian failure by mesenchymal stem cells in rats; Approval document of Ethics Committee: Qilu Hospital of Shandong university; Approval No.: 2021-147; IACUC Issue No.: DWLL-2021-147; Approval date: December 15, 2021).
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
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 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-nc-nd/4.0/.
About this article
Cite this article
Zhang, Y., Li, D., Han, Y. et al. Intraovarian injection of 3D-MSC-EVs-ECM gel significantly improved rat ovarian function after chemotherapy. Reprod Biol Endocrinol 22, 125 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12958-024-01299-3
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12958-024-01299-3