Ethics statement
The experiments were performed in accordance with the Guide for the Care and Use of Agricultural Animals in Agricultural Research and Teaching, and all procedures were approved by the Institutional Animal Care and Use Committee at the China Agricultural University (Beijing, China). Some publicly available data was analyzed to support our work. The endometrial proteomes of pregnant and pregnancy failed ewes during the peri-implantation period were obtained from Haichao Zhao et al. [42]. Gene expression in endometrial samples from women with and without endometriosis was obtained from GSE135485. The ChIP-seq data of histone lactylation were from GSE115354 [58]. The RNA-seq data of the endometrial luminal epithelium, endometrial glandular epithelium, and conceptus from ewes on day of pregnancy 12, 14, 16 and 20 were from GSE87017 [45]. The RNA-seq data of the pre-receptive to receptive human endometrium were from S. Hu, et al. [71].
Animals and treatment.
Chinese Small Tail Han ewes with normal estrous cycles were selected for the present study. The procedures of estrous synchronization, superovulation, artificial insemination (AI), and transfer of good-quality blastocysts were performed as described in our previous study [42].
ICR female mice aged 7–8 week and ICR male mice aged 10–12 week were fed ad libitum
and housed under controlled lighting conditions (12 light:12 dark). They were maintained under specific pathogen-free conditions. All animal experiments were approved by and performed in accordance with the guidelines of the Institutional Animal Care and Use Committee of China Agricultural University.
Sample collection
We collected good-quality embryos from thirty donors at day 6.5 of pregnancy. Then, two well-developed blastocysts were transferred into each synchronized recipient ewe (forty-eight synchronized ewes). Sampling procedures were similar to the methods detailed in our previous study [42]. Briefly, all recipients were slaughtered at day 17 of pregnancy, then their uteri were collected and the conceptuses were flushed out using phosphate-buffered saline (PBS). Thirty-seven recipients had filamentous conceptuses. The endometrial caruncular (C) areas and intercaruncular (IC) areas were collected and processed as described by Attia et al. [21]. Opening the ipsilateral uterine horn longitudinally by scissors, the C areas were carefully cut out and collected, and then the IC areas were sampled. These samples were stored at liquid nitrogen until further analysis (Fig. 1A).
Protein extraction
We divided thirty-six samples into three equally pools, with twelve samples in each pool (Fig. 1A). Each pool was ground to powder in liquid nitrogen and stored overnight at − 20 °C after adding a five-fold volume of chilled acetone containing 10% trichloroacetic acid (TCA) and 10 mmol/L dithiothreitol (DTT). The samples were then centrifuged at 4°C, 16,000 × g for 20 min and the supernatant was discarded. The precipitates were mixed with 1 mL of chilled acetone containing 10 mmol/L DTT, stored for 30 min at − 20°C, and centrifuged at 4°C, 20,000 × g for 30 min. Centrifugation was repeated several times until the supernatant was colorless. The pellets were air-dried, dissolved in lysis buffer containing 1 mmol/L phenylmethanesulfonyl fluoride (PMSF), 2 mmol/L ethylenediaminetetraacetic acid (EDTA), and 10 mmol/L DTT and sonicated at 200 W for 15 min before being centrifuged at 30,000 × g at room temperature for 30 min. The protein concentration in the supernatant was then detected by using the Bradford method.
Peptide digestion
Proteins (50 µg) were taken from each sample, and isopycnic samples were prepared by adding 8 mol/L urea solution. To reduce disulfide bonds, the samples were incubated with 10 mmol/L DTT at 56°C for 1 h, and then cysteine bonding was blocked using 55 mmol/L iodoacetamide (IAM) in a dark room for 45 min. Thereafter, each sample was diluted 8-fold with 50 mmol/L ammonium bicarbonate and digested with Trypsin Gold at a protein: trypsin ratio of 20:1 at 37°C for 16 h. Following desalting using a Strata X C18 column (Phenomenex, Torrance, CA, USA), the samples were vacuum dried. Peptides generated from digestion were directly loaded for liquid chromatography-electrospray ionization tandem mass spectroscopy (LC-ESI-MS/MS) analysis.
LC-ESI-MS/MS analysis with a linear ion trap-orbitrap (LTQ-orbitrap) collision induced dissociation (CID)
Each sample was resuspended in buffer A [2% acetonitrile (ACN), 0.1% formic acid (FA)] and centrifuged at 20,000 × g for 10 min. The final peptide concentration for each sample was approximately 0.5 µg/mL. The digested samples were fractionated using a Shimadzu LC-20AD nano-high performance liquid chromatography (HPLC) system (Shimadzu, Kyoto, Japan). Each sample (10 µL) was loaded by the autosampler onto a 2 cm C18 trap column (200 µm inner diameter), and the peptides were eluted onto a resolving 10 cm analytical C18 column (75 µm inner diameter) prepared in-house. The samples were loaded at a flow rate of 15 µL/min for 4 min, and then a 91 min gradient from 2–35% buffer B (98% ACN, 0.1% FA) was run at a flow rate of 400 nL/min, followed by a 5 min linear gradient to 80% buffer B that was maintained for 8 min before finally returning to 2% buffer B within 2 min. The peptides were subjected to nano-electrospray ionization and then detected by MS/MS in an LTQ Orbitrap Velos (Thermo Fisher Scientific, Bremen, Germany) coupled online to an HPLC system. Intact peptides were detected in the Orbitrap analyzer at a resolution of 60,000 m/z. Peptides were selected for MS/MS using the CID operating mode with a normalized collision energy setting of 35%, and ion fragments were detected in the LTQ. One MS scan followed by ten MS/MS scans was applied for the ten most abundant precursor ions above a threshold ion count of 5,000 in the MS survey scan. Dynamic exclusion was used, with the following parameters: Repeat counts = 2; repeat duration = 30 s; and exclusion duration = 120 s. The applied electrospray voltage was 1.5 kV. Automatic gain control (AGC) was used to prevent overfilling of the ion trap; 1 × 104 ions were accumulated in the ion trap to generate CID spectra. For MS scans, the m/z scan range was 350 to 2,000 Da.
Proteomic analysis
MaxQuant software (version 1.1.1.36) was used to analyze the mass spectra. Bos taurus is the most robust and extensive protein annotated species with a genomic database, with a strong homology to sheep [75, 87]. Therefore, we generated one reference protein database by integrating the following databases and sequences of cattle proteins and limited publicly available sheep proteins, and removed duplicate proteins: GenBank nr (20110403), UniProt cow proteins (20110503), sheep proteins (http://www.livestockgenomics.csiro.au/sheep/), and cow proteins (http://genomes.arc.georgetown.edu/drupal/bovine/). The MS/MS data were searched against the reference protein database using the search engine embedded in MaxQuant. Up to two missed cleavages were allowed. The first search was set to 20 ppm, and the MS/MS tolerance for CID was set to 0.5 Da. The false discovery rate (FDR) was set to 0.01 for peptide and protein identifications, which was estimated based on the fraction of reverse protein hits [88, 89]. Proteins were considered identified when at least two peptides were identified, at least one of which was uniquely assignable to the corresponding sequence. In the case of identified peptides that were all shared between two proteins, these were combined and reported as one protein group. To control the false match frequency, the contents of the protein table were filtered to eliminate identifications from the reverse database and common contaminants [90, 91]. The minimum peptide length was set to six amino acids. To perform label-free quantification analysis, the MaxQuant software suite containing an algorithm based on the extracted ion currents (XICs) of the peptides was used. Xcalibur 2.1 (Thermo Scientific) was used as quality control program to check the quality of chromatographs. This specific label-free processing method was performed as described by Waanders et al. [92].
Cell culture
A human endometrial cancer cell line (Ishikawa, ATCC, Manassas, VA, USA). Ishikawa cells were grown at 37°C in DMEM/F-12, HEPES (Gbico, UK) supplemented with 10% fetal bovine serum (FBS; Hyclone, Logan, UT, USA) and 1% penicillin/streptomycin (Invitrogen, Waltham, MA, USA) in a humidified 5% CO2 incubator.
Western blotting
Samples of acid extracted histones from IC areas, C areas, and Ishikawa cells were fractionated with 15% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to polyvinylidene fluoride membranes. The membranes were blocked with 5% skimmed milk in Tris buffered saline-Tween for one hour, followed by probing with anti-H3K18la (1:2000, PTM-1406, PTM Bio Inc., Hangzhou, China), anti-H3 (1:1000, ab12079, Abcam, Cambridge, MA, USA), anti-CLDN4 (1:500, sc-376643, Santa Cruz Biotechnology), or anti-ALB (1:500, sc-271605, Santa Cruz Biotechnology). After incubation at 4°C overnight, the membrane was probed with horseradish peroxidase-conjugated secondary antibody for one hour. After washing the blots were visualized using the ECL system (Bio-Rad, Hercules, CA, USA). The grey level of each bands was calculated by ImageJ (https://imagej.nih.gov/ij/, NIH, Bethesda, MD, USA).
The detection of lactate concentration
The lactate concentration in tissues was detected using a lactate assay Kit (MAK064, Sigma, St. Louis, MO, USA). The total protein concentration was quantified using an enhanced BCA protein assay kit (Beyotime Biotechnology, Jiangsu, China). Finally, the lactate concentration was normalized by the total protein concentration.
The detection of GSH/GSSG
Cells were harvested and washed three times with PBS. Then, the concentrations of GSH and GSSG were quantified using a GSH and GSSG Assay Kit following the manufacturer’s instruction (Beyotime Institute of Biotechnology, Nanjing, China). Each experiment was performed with three replicates.
Measurement of ROS
The intracellular ROS levels of Ishikawa cells were determined using a Reactive Oxygen Species Assay Kit (Beyotime Institute of Biotechnology), according to the manufacturer’s instruction. The fluorescence signals were imaged using a confocal laser scanning microscope (Digital Eclipse C1; Nikon). The fluorescent intensity was calculated using ImageJ.
Cell viability assay
Ishikawa cells were resuspended and seeded into 96-well culture plates at a density of 2,000 cells per well. Then cells were treated with 10 mM sodium L-lactate for 12 hours and 24 hours. Cell proliferation was detected using an Enhanced Cell Counting Kit-8 (CCK-8) (Beyotime).
The detection of cell apoptosis
A terminal deoxynucleotidyl transferase mediated dUTP nick end-labeling (TUNEL) assay was used to analyze cell apoptosis, following the manufacturer’s instruction of the In Situ Cell Death Detection Kit (11684817910, Roche Molecular Biochemicals, Indianapolis, IN, USA). Ishikawa cells were seeded into 24-well culture plates, and treated with 10 mM, 50 mM, or 100 mM sodium L-lactate for 24 hours. The fluorescence signals were imaged using a confocal laser scanning microscope (Digital Eclipse C1; Nikon). The fluorescent intensity was calculated using ImageJ.
Cell adhesion assay
Fibronectin (10 µg/mL, Sigma) was used to coat a 35-mm culture dish, overnight at 4°C, and then washed three times with PBS. Ishikawa cells were resuspended in serum-free medium (DMEM/F-12, HEPES (Gbico, UK) supplemented with 1% penicillin/streptomycin (Invitrogen)) and 5 µg/mL, 25 µg/mL, 100 µg/mL BSA (Bovine serum albumin, Sigma) and then plated in the 35-mm dishes. After 2 hours of incubation, the non-adherent cell containing media was aspirated off and each well was washed gently with PBS. Then, images of adherent cells were acquired under a microscope (Olympus) from at least ten random fields.
Preparation of mouse embryos
All experiments involved in embryo preparation were performed as previously described [93], with minor modifications. ICR female mice were superovulated by intraperitoneal injection of 5 IU pregnant mare serum gonadotropin and a further intraperitoneal injection 48 h later of 5 IU human chorionic gonadotropin (hCG). Then sperm were obtained from the cauda epididymis and capacitated for 1 h in human tubal fluid (HTF; SAGE) medium at 37°C in 5% CO2. Oocytes were collected from the ampullae at 14 h after hCG treatment. Gametes were then co-incubated in HTF medium for 4 h at 37°C in 5% CO2. After 4 h in the incubator, zygotes were washed and cultured to the blastocyst stage in potassium simplex optimization medium containing amino acids (KSOM + AA; Millipore) under mineral oil at 37°C in 5% CO2.
In vitro embryo implantation model and mouse embryo attachment assay
The in vitro embryo implantation model was constructed according to the previous reports [59, 60]. For attachment assay, well-developed mouse blastocysts were collected and then transferred onto differently treated Ishikawa cells in serum-free medium, with ten blastocysts per well. After the blastocysts and Ishikawa cells were cocultured for 24 or 48 hours, a standardized plate movement protocol was implemented to measure the number of attached embryos [94, 95].
Fluorescent immunocytochemistry
Ishikawa cells were co-cultured with blastocysts 72 hours, then fixed with 4% paraformaldehyde for further detection of H3K18la. For albumin overload assay in Fig. 2C and Fig. S5B, control group was incubated 30 min with FBS-medium (DMEM/F-12, HEPES supplemented with 1% penicillin/streptomycin and 10% FBS), while MSA group were incubated with MS-medium (DMEM/F-12, HEPES (Gbico, UK) supplemented with 1% penicillin/streptomycin (Invitrogen)) and 10% mouse serum) 30 min, then fixed with 4% paraformaldehyde for further detection of ALB and CLDN4. Immunostaining was performed according to standard protocols using the following primary antibodies: anti-H3K18la (1:1000, PTM-1406, PTM Bio Inc.), anti-ALB (1:250, sc-271605, Santa Cruz Biotechnology), anti-CLDN4 (1:250, 16195-1-AP, Proteintech). And appropriate Alexa Fluor dye conjugated secondary antibodies (Invitrogen) were used. Nuclei were stained with DAPI (Life Technologies). The fluorescence signals were imaged using a confocal laser scanning microscope (Digital Eclipse C1; Nikon). Data analysis was performed by ImageJ.
Intrauterine injection
ICR female mice were superovulated by intraperitoneal injection of 5 IU pregnant mare serum gonadotropin and a further intraperitoneal injection 48 h later of 5 IU human chorionic gonadotropin (hCG). Each female mouse was caged with one male and allowed to mate naturally overnight. Day 1 of pregnancy was designated as the next morning when a vaginal plug was formed. On day 4 of pregnancy, each uterine horn of female mice was slowly injected with 5 µL of saline (or 10 mg/mL, 25 mg/mL oxamate, or 10 mg/mL oxamate with 0.04 mM, 0.2 mM, 1mM, 5mM lactate) using a 26-gauge Hamilton syringe (no. A6410, Sigma). In the morning of day 5, the treated animals were sacrificed to count the numbers of implantation sites [96].
Analyses of differentially abundant proteins
To facilitate data analysis, all proteins were mapped to the Ensembl Bos Taurus gene ID. P values from student’s t-test were corrected for multiple hypothesis tests using the false discovery rate (FDR) procedure [97]. For each comparison, gene expression levels were considered significantly different when FDR < 0.05 and fold change (FC) > 2. The protein quantification values of the conceptus, C area, IC area, and DAPs of each comparison (conceptus vs. C area and conceptus vs. IC area) are shown in Table. S1.
Annotations of differentially abundant membrane and secreted proteins
The annotations of membrane and secreted proteins were processed as described by Vento-Tormo et al [35]. Briefly, DAPs were mapped to UniProt (https://www.uniprot.org/), then KW-0964 (secreted) was used to screen out the secreted partners. KW-1003 (cell membrane) was used to screen out the plasma membrane proteins. Peripheral proteins from the plasma membrane were annotated using the UniProt Keyword SL-9903, and the remaining proteins were annotated as membrane proteins, which act as extracellular signal receptors. Interestingly, some proteins were annotated as both secreted proteins and membrane proteins, such as heat shock protein 90 alpha family class B member 1 (HSP90AB1) and elastin microfibril interfacer 1 (EMILIN1). The differentially abundant secreted proteins or membrane proteins are shown in Table S2. Phenotype annotations of differentially abundant membrane proteins or secreted proteins were analyzed based on the MGI database (Mouse Genome Informatics, http://www.informatics.jax.org/phenotypes.shtml).
Construction of membrane-secreted partner interactions
We used the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING version 11.0; https://string-db.org/ [98]) to build the membrane-secreted partner interactions using edge information from three separate forms of evidence: Databases, experiments, and text mining. Firstly, we inputted a secreted protein (or a membrane protein) to acquire its interacting partners and interaction scores. Then we mapped its interacting partners to UniProt to screen out the membrane partners (or secreted partners). Finally, we chose the membrane partner (or secreted partner) with the highest interaction score. In this way, we constructed the interactions of differentially abundant membrane proteins (or secreted proteins) with their secreted partners (or membrane partners). All the interactions are shown in Table S3.
Gene ontology (GO) and KEGG pathway analysis and pathway network construction
DAVID version 6.8 (https://david.ncifcrf.gov/home.jsp) enables the generation of specific functional annotations of biological processes affected by treatment from the target gene lists produced in high-throughput experiments [99, 100]. We used DAVID to carry the gene-annotation enrichment analysis to obtain a functional view of the DAPs. The genetic background in DAVID is Bos Taurus, which was a default parameter given by DAVID when we uploaded the DAPs list. Visualizations of significant GO and Kyoto Encyclopedia of Genes and Genomes (KEGG) categories (P values < 0.05) was carried by the “ggplot2” package [101] in R (R version 3.5.1. https://www.R-project.org/.). The pathway network construction and key pathway findings were determined using PathwayConnector (PC) (http://bioinformatics.cing.ac.cy/PathwayConnector/#) [47].
Hierarchical clustering analysis, Principal Components Analysis (PCA), Gene Set Enrichment Analysis (GSEA), and protein-protein network construction
Unsupervised hierarchical clustering analysis was carried using the “hclust” function in R. PCA of all samples was carried using the “Prcomp” function in R. Significantly differentially regulated pathways were identified by GSEA (NES > 1, FDR < 0.25) [102, 103]. We used STRING to construct the protein-protein interaction network of DAPs, and then visualized using the cytoHubba plugin in the Cytoscape software according to the degree number [104].
Constructed the protein-protein docking model of ALB-CLDN4
The crystal structure of ALB (1AO6) and CLDN4 (7KP4) was downloaded from protein data bank (http://www.rcsb.org/). And the ClusPro (https://cluspro.org) was used for protein-protein docking [105–108]. We chose the balanced pattern to predict the interaction of ALB and CLDN4, and cluster scores were used to evaluated the docking model. Then the PyMOL software was used to show the docking model with highest score as cartoon and surface shapes.
Co-immunoprecipitation assay
Samples were fractionated using RIPA (beyotime) supplemented with protease and phosphatase inhibitors (beyotime), and used for immunoprecipitation using Dynabeads protein G according to manufacturers’ protocol (10007D, Life technologies). CLDN4 (1 µg / 1 mg lysate, 16195-1-AP, Proteintech) and IgG antibody (rabbit anti-mouse, 06-371, Sigma) were used for immunoprecipitation. At the final step, the beads-antibody-antigen complexes were eluted in 50 µL loading buffer (2 × Laemmli Sample Buffer (Bio-Rad), 5% 2-Mercaptoethanol (Sigma)), and boiled at 70°C for 10 min. The beads were separated from the magnet and supernatant (containing the eluted protein) and used for western blotting.
Real-time quantitative PCR analysis
Total RNAs from the mouse E6.5 embryo and decidua tissues were extracted using TRIzol reagent (Invitrogen). cRNA was reverse-transcribed into complementary DNA (cDNA) with a HiScript II reverse transcriptase reagent kit (Vazyme, Nanjing, China). Real-time PCR was performed using SsoFast EvaGreen Supermix (BioRad) in a CFX96 real-time PCR machine (Bio-Rad). Actb was used as internal reference. At least three independent experiments were performed. Primers used for qPCR: Actb-F: TGGCGCTTTTGACTCAGGAT, Actb-R: GGGATGTTTGCTCCAACCAA; Alb-F: GAAAACCAGGCGACTATCTCCA; Alb-R: TGCACACTTCCTGGTCCTCA; Cldn4-F: CGCTACTCTTGCCATTACGC, Cldn4-R: TCACTCAGCACACCATGACTTG.
Fluorescent immunohistochemistry
Fixed E6.5 tissues were prepared according to standard protocols for paraffin embedding. The paraffin-embedded tissues were sectioned serially at 5 µm thick. The sections were microwaved in the antigen unmasking solution, blocked with bovine serum albumin solution, and incubated overnight with anti-ALB (1:200, sc-271605, Santa Cruz Biotechnology) and anti-CLDN4 (1:200, 16195-1-AP, Proteintech) at 4°C. In the following day, slides were washed in PBS containing 0.1% Tween 20 (PBS-T), incubated with secondary antibodies conjugated with Alexa Fluor 488 (anti-mouse; Invitrogen) or Alexa Fluor 594 (anti-rabbit; Invitrogen) for 1 h at room temperature, washed with PBS-T, counterstained with DAPI and mounted. The fluorescence signals were imaged using a confocal laser scanning microscope (Digital Eclipse C1; Nikon). Data analysis was performed by ImageJ.
Statistical analysis
The P value of Student’s t-test was calculated using GraphPad Prism 7.0 software (GraphPad Inc., La Jolla, CA, USA) or R for individual analysis. It was considered significant when the P value < 0.05. Error bars represent the means ± SEM (Standard Error of the Mean). Details of individual tests are outlined within each figure legend.