Establishment of Animal and Cell Models
Husbandry of C57BL/6J Mice
In this study, male C57BL/6J mice (Strain number N000013), aged 5–8 weeks, were procured from GemPharmatech (Nanjing, China). Upon arrival, the mice were housed in a specific pathogen-free (SPF) environment, where the ambient temperature was maintained at 22 ± 2°C with a relative humidity of 50 ± 10%, adhering to a 12-hour light/dark cycle. The mice were kept in standard ventilated cages, with 3–5 mice per cage. Access to food and water was provided ad libitum. Throughout the experimental period, the welfare and health status of the animals were routinely monitored by a professional team. The Animal Experimental Ethics Committee of Southeast University approved these experiments (Approval number 20200226001), which were conducted in compliance with Chinese legislation concerning the use of experimental animals.
Establishment of the ARDS Mouse Model
An ARDS mouse model was established using a chemical induction method. Male C57BL/6J mice, aged 5–8 weeks and weighing between 20–25 grams, were used for the experiments. The mice were acclimatized for at least one week prior to the experiment. For the modeling procedure, the mice were first anesthetized with 2%-3% isoflurane inhalation. Subsequently, tracheal intubation was performed, followed by the intratracheal injection of 5 mg/kg body weight of LPS (Beyotime, cat. no. ST1470) to establish the ARDS model. After the establishment of the model, the mice were returned to clean cages for recovery. Over the next 24 hours, the development of ARDS was assessed by monitoring parameters such as respiratory rate, changes in body weight, and behavioral activity.
Intravenous Injection of MSCs in Mice
MSCs were administered to the experimental mice via tail vein injection. Initially, C57BL/6J mouse bone marrow-derived MSCs (OriCell, cat. no. MUBMX-01001) were purchased. The experimental mice were fasted a day before the MSCs injection, while ensuring an adequate supply of water. On the day of injection, after establishing the ARDS mouse model and 4 hours post-modeling, the mice were secured on a specialized tail vein injection stand. MSCs were diluted in sterile saline to a concentration of 1×10^6 cells/200 µL and then slowly injected into the mouse tail vein. Care was taken to avoid the formation of air bubbles during the injection. After the injection, the mice were returned to their cages for recovery, with close observation to ensure no apparent discomfort or abnormal behavior.
Culture, Passaging, and Cryopreservation of MSCs
Bone marrow-derived MSCs from C57BL/6J mice were expanded in vitro. The culture medium consisted of DMEM/F-12 (Gibco, cat. no. 11320033), 10% fetal bovine serum (Gibco, cat. no. 10099141C), and 1% penicillin-streptomycin (Gibco, cat. no. 15140148). Cells were cultured in a humidified incubator at 37°C with 5% CO2. When cells reached 80–90% confluency, passaging was performed.
For passaging, cells were first washed twice with PBS (Procell, cat. no. PB180327) and then treated with 1 mL of 0.25% trypsin-EDTA solution (Gibco, cat. no. 25200114) in a 25T flask. Cells were incubated at 37°C for 1–2 minutes until detachment from the flask bottom. Immediately, an equal volume of complete culture medium was added to neutralize trypsin activity. The cell suspension was transferred to a 15 mL centrifuge tube and centrifuged at 300×g for 5 minutes. The supernatant was discarded, and the cells were resuspended in fresh culture medium and seeded into new flasks for continued cultivation.
When MSCs reached the appropriate passage number for cryopreservation, they were first collected following the passaging steps. Cells were resuspended at a concentration of 1×10^6 cells/mL in cryopreservation solution (NCM Biotech, cat. no. C40100). The cell suspension was aliquoted into pre-chilled cryovials, which were then placed in a cryogenic box in a -80°C freezer overnight before transferring to liquid nitrogen for long-term storage.
Culture, Passaging, and Cryopreservation of MPMECs
We cultured an immortalized mouse pulmonary microvascular endothelial cell line (MPMECs), whose proliferative capacity, morphological characteristics, genetic stability, and expression of endothelial cell markers have been confirmed in previous studies[24]. The culture medium comprised of basic DMEM/F-12, 5% fetal bovine serum, 1% endothelial cell growth supplement (ScienCell, cat. no. 1052), 1% penicillin-streptomycin, 90U/mL heparin (Sigma-Aldrich, cat. no. H3149), and 92mg/L D-valine (Sigma-Aldrich, cat. no. V1255). Cells were maintained in a humidified incubator at 37°C with 5% CO2, with fresh medium replacement every 1–2 days. Passaging was performed when cells reached 70%-80% confluence.
For passaging MPMECs, cells were first washed twice with PBS and then treated with 1mL of 0.25% trypsin-EDTA solution diluted tenfold in PBS. Cells were incubated at 37°C for 1–2 minutes until detachment from the flask bottom. Subsequently, an equal volume of culture medium containing FBS was added to neutralize trypsin. The cell suspension was transferred to a centrifuge tube and centrifuged at 300×g for 5 minutes. The supernatant was discarded, and the cells were resuspended in fresh culture medium and seeded into new flasks for continued cultivation.
For cryopreservation of MPMECs, cells were first collected following the passaging protocol. Cells were resuspended at a concentration of 1×10^6 cells/mL in cryopreservation solution. The cell suspension was aliquoted into pre-chilled cryovials. The vials were then placed in a cryogenic box in a -80°C freezer overnight before transferring to liquid nitrogen for long-term storage.
Mitochondrial Damage Induction with Rotenone
In this study, rotenone (MedChemExpress, cat. no. HY-B1756) was employed to induce mitochondrial damage, simulating conditions of mitochondrial dysfunction in ARDS. Rotenone is a naturally occurring compound known to impair mitochondrial respiration by Complex I of the mitochondrial electron transport chain. Cells intended for the experiment were cultured to an appropriate density at 37°C and 5% CO2 prior to treatment. A working solution of rotenone was prepared. The rotenone powder was first dissolved in dimethyl sulfoxide (DMSO) (MedChemExpress, cat. no. HY-Y0320) to create a high-concentration stock solution (1 mM). For experimental use, this stock solution was further diluted to the desired final concentration (100nM) in the culture medium. During treatment, the culture medium containing rotenone was directly added to the cell culture dishes, gently swirled to ensure uniform mixing. The cells were stimulated for 4 hours. After the treatment, the medium containing rotenone was removed, and the cells were washed with PBS, followed by replacement with fresh culture medium for subsequent experiments.
Method Details
Flow Cytometry Analysis of Mitochondrial Transfer from MSCs to MPMECs
MSCs and MPMECs were cultured separately until reaching 70–80% confluency, and mitochondria in MSCs were labeled with MitoTracker Deep Red FM (Invitrogen, cat. no. M22426). Briefly, MitoTracker Deep Red FM was dissolved in DMSO to make a 1 mM stock solution and then diluted with complete culture medium to a working concentration of 200 nM (2 µL of stock solution added to 10 mL of complete culture medium). The culture medium was removed from the flasks, replaced with pre-warmed working solution, and the cells were incubated for 30 minutes. Afterward, the cells were washed with PBS and fresh culture medium was added.
For co-culture experiments, 1×10^5 mitochondria-prelabeled MSCs were co-cultured with MPMECs for 24 hours. Post co-culture, the cells were treated with 0.25% trypsin-EDTA for digestion, then collected by centrifugation and washed with PBS. Prior to flow cytometry, CD31 antibody (BD Pharmingen, cat. no. 553373) was used to specifically label surface proteins of MPMECs. Dual labeling with MitoTracker Deep Red FM and CD31 antibody allowed for the distinction between the two cell types in flow cytometry and the detection of MSCs' mitochondria in MPMECs.
Flow Cytometry Analysis of Mitochondrial Transfer from MSCs to MPMECs in ARDS Mouse Model
An ARDS mouse model was established, and MSCs were cultured to 70–80% confluence and labeled with MitoTracker Deep Red FM for mitochondrial tracking. Four hours after establishing the ARDS mouse model, these mitochondria-prelabeled MSCs were administered via tail vein injection to the ARDS model mice. Twenty-four hours later, the mice were euthanized, and their lung tissues were rapidly harvested. Under sterile conditions, the lung tissues were subjected to mechanical and enzymatic digestion to prepare a single-cell suspension, which was then collected by centrifugation and washed with PBS.
MPMECs were specifically labeled using a fluorescently-tagged CD31 antibody, which targets unique surface proteins of these cells. Flow cytometry was employed to distinguish between endothelial and non-endothelial cells, and to detect the presence of MSC mitochondria within MPMECs. The fluorescence intensity of MitoTracker Deep Red FM within the MPMEC population and the proportion of cells were analyzed to assess the degree of mitochondrial transfer from MSCs to MPMECs.
Immunofluorescence Staining of Mouse Lung Tissue to Detect Mitochondrial Transfer from MSCs to MPMECs in ARDS
Mitochondria of in vitro cultured MSCs were labeled with MitoTracker dye (MitoTracker Deep Red FM). Four hours post-establishment of the ARDS mouse model, these pre-labeled mitochondria MSCs were injected into the ARDS model mice via tail vein. Twenty-four hours after ARDS modeling, the mice were euthanized, and their lung tissues were rapidly extracted and immediately frozen at -80°C. Frozen sections were then prepared, slicing the lung tissues into 5 µm thick sections.
The lung tissue sections underwent immunofluorescence staining, with primary antibody incubation using a CD31 antibody (Abcam, cat. no. ab182981) for labeling MPMEC surface markers. Excess primary antibody was washed off, followed by incubation with a secondary antibody, Alexa Fluor 488 (Abcam, cat. no. ab150077), for specific labeling of MPMECs. Finally, the sections were observed and imaged using a fluorescence microscope. The co-localization of MSCs' MitoTracker labeling (red) with MPMECs' CD31 labeling (green) was analyzed to determine whether MSC mitochondria were transferred to MPMECs.
Confocal Microscopy Observation of Mitochondrial Transfer and Tunneling Nanotubes Between MSCs and MPMECs
Initially, MSC mitochondria were labeled using MitoTracker Deep Red FM. MPMEC mitochondria were labeled with MitoTracker Green FM (Invitrogen, cat. no. M7514). Briefly, MPMECs were cultured in confocal dishes to 50% confluency. MitoTracker Green FM was dissolved in DMSO to make a 1 mM stock solution and then diluted with complete culture medium to a working concentration of 100 nM (1 µL of stock solution added to 10 mL of complete culture medium). The culture medium was removed from the confocal dishes, replaced with pre-warmed working solution, and the cells were incubated for 30 minutes. Subsequently, the cells were washed with PBS and fresh culture medium was added. Mitochondria-labeled MSCs were then added to the confocal dishes containing MPMECs and co-cultured for 12 hours. During this period, MSCs and MPMECs might exchange mitochondria via tunneling nanotubes. Under the confocal microscope, the co-localization of MSC mitochondria (red fluorescence) and MPMEC mitochondria (green fluorescence) was observed, along with the morphology of tunneling nanotubes.
Fluorescence Microscopy Observation of Mitochondrial Transfer from MSCs to MPMECs in Isolated Culture
In this study, we employed a Transwell system to isolate and culture MSCs and MPMECs, and observed the mitochondrial transfer from MSCs to MPMECs using fluorescence microscopy. Initially, MPMEC mitochondria were labeled with MitoTracker Deep Red FM, and MPMECs were cultured in the lower chamber of the Transwell system. MSC mitochondria were labeled with MitoTracker Green FM, and these pre-labeled MSCs were seeded in the upper chamber of the Transwell system. After 24 hours of isolated co-culture, allowing for the transfer of mitochondria from MSCs to MPMECs, fluorescence microscopy was performed. Appropriate fluorescence excitation and emission wavelengths were adjusted to observe the MPMECs in the lower layer of the Transwell system. The co-localization of red and green fluorescence within MPMECs confirmed the transfer of mitochondria from MSCs to MPMECs during isolated culture.
Preparation of Single-Cell Suspension from Mouse Lung Tissue
Initially, mice were euthanized following full anesthesia, and the lungs were rapidly excised and placed in pre-cooled PBS. Under sterile conditions, the lung tissues were thoroughly washed in PBS to remove blood. The washed tissues were then transferred to a sterile cutting board, where they were minced into 1–2 mm pieces using surgical scissors and tweezers. These tissue pieces were transferred to a digestion buffer containing collagenase (Beyotime, cat. no. ST2303) and DNase (Beyotime, cat. no. D7073), and incubated at 37°C with gentle shaking for 1–2 hours to dissociate the tissue and release individual cells.
Following digestion, the mixture was filtered through a 100 µm cell strainer to remove undigested tissue fragments. The collected cell suspension was then centrifuged (300×g for 5 minutes), and the supernatant was discarded. Subsequently, the cell suspension was treated with red blood cell lysis buffer (Beyotime, cat. no. C3702) to eliminate red blood cells. After 5 minutes, the suspension was centrifuged again, the supernatant was discarded, and the cells were resuspended in PBS warmed to room temperature.
Western Blot Analysis of Mitochondrial Complex I Expression
Initially, total protein was extracted from the target cells or tissues, and the protein concentration was determined using a BCA Protein Assay Kit (Beyotime, cat. no. P0010). The protein samples were then mixed with SDS loading buffer and boiled at 95°C for 10 minutes to denature the proteins. Subsequently, the samples (15 µg) were loaded onto an SDS-PAGE gel for electrophoretic separation. After electrophoresis, the proteins were transferred to a PVDF membrane, which was then blocked with 5% BSA solution to prevent non-specific binding, and incubated at room temperature for 1 hour. The membrane was then incubated overnight at 4°C with a specific primary antibody against mitochondrial Complex I (Abcam, cat. no. ab110245).
The next day, the membrane was washed three times with TBS containing 0.1% Tween-20 for 10 minutes each to remove excess primary antibody. This was followed by incubation with the corresponding HRP-conjugated secondary antibody (Beyotime, cat. no. A0208) at room temperature for 1–2 hours. After the incubation with the secondary antibody, the membrane was washed again and then treated with chemiluminescent substrate to detect the protein signal. To ensure consistency in protein loading, the membrane was also probed with an antibody against the internal control protein β-actin (Beyotime, cat. no. AF0003).
Flow Cytometry Analysis of CFSE Mean Fluorescence Intensity in MPMECs
In this study, flow cytometry was utilized to detect CFSE staining in MPMECs to evaluate cell proliferation and division. Initially, MPMECs were cultured to 50% confluency, and CFSE (Invitrogen, cat. no. C34570) was added to the culture at a concentration of 5 µM. The cells were then incubated at 37°C with 5% CO2 for 15–20 minutes. During this period, CFSE permeated the cell membrane and bound to intracellular amino acids, forming a stable fluorescent complex. After incubation, cells were washed with PBS and resupplied with complete culture medium. Subsequently, 1×10^5 MSCs were added to the MPMEC culture system according to the experimental groups, and after 24 hours of co-culture, the treated cells were collected, washed with PBS, labeled with CD31 antibody to mark MPMECs, and prepared for flow cytometry analysis. In the flow cytometer, appropriate lasers and detectors were set to differentiate MPMECs while simultaneously measuring the mean fluorescence intensity of CFSE in MPMECs. Data were analyzed using FlowJo software, and cell proliferation was assessed by calculating the mean fluorescence intensity of CFSE.
Flow Cytometry Analysis of ROS Mean Fluorescence Intensity in MPMECs
Flow cytometry was employed to assess the levels of reactive oxygen species (ROS) in MPMECs. MPMECs were cultured to 50% confluency and subjected to cell treatments according to experimental groups, with the MSC-treated experimental group receiving 1×10^5 MSCs. After 24 hours of co-culture with MSCs, the cells were collected. A specific ROS assay kit (Elabscience, cat. no. E-BC-K138-F) was used to treat the cell samples. Briefly, the ROS probe DCFDA was used for ROS detection, dissolving DCFDA at 10 µM in serum-free culture medium, and incubating MPMECs with this solution for approximately 20–30 minutes at 37°C. During incubation, DCFDA penetrated the cells and was converted into a fluorescent compound under the influence of ROS. After incubation, cells were washed with PBS to remove uninternalized or unreacted DCFDA. Subsequently, cells were labeled and distinguished as MPMECs using a CD31 antibody. Flow cytometry analysis was performed to analyze the fluorescence signal, detecting and assessing the mean fluorescence intensity of ROS in CD31-positive cells.
Flow Cytometry Analysis of Cell Apoptosis in MPMECs
Flow cytometry was utilized to assess apoptosis in MPMECs. Prior to the experiment, MPMECs were cultured to an appropriate density and subjected to experimental treatments, with 1×10^5 MSCs added to the MSC-treated group. After cell treatment, an Annexin V/PI Apoptosis Detection Kit (Elabscience, cat. no. E-CK-A211) was used for labeling and analysis. Firstly, the treated cells were collected and washed with PBS to remove serum and dead cells from the culture medium. The cell concentration was adjusted to 1×10^6 cells/mL, and then cells were resuspended in staining buffer containing Annexin V-FITC and PI, gently mixed to avoid excessive agitation. The cells were incubated in the dark at room temperature for 15–20 minutes. Following staining, MPMECs were labeled with CD31, and then analyzed using flow cytometry.
Using flow cytometry software, CD31 positive MPMECs were selected, and cells were categorized into early apoptotic cells (Annexin V positive/PI negative), late apoptotic or necrotic cells (Annexin V positive/PI positive), and viable cells (Annexin V negative/PI negative). Based on these classifications, the level of apoptosis in MPMECs could be quantitatively analyzed.
Hematoxylin and Eosin (H&E) Staining and Lung Injury Scoring of Mouse Lung Tissue
In this study, mouse lung tissues were stained with Hematoxylin and Eosin (H&E) to assess the extent of lung injury. Firstly, after experimental treatment, mice were euthanized, and lung tissues were rapidly excised. The tissues were fixed in 4% paraformaldehyde solution for at least 24 hours to preserve the integrity of the tissue structure. After fixation, the lung tissues underwent dehydration, clearing, and paraffin infiltration, followed by embedding in paraffin blocks. The paraffin-embedded lung tissues were sectioned into 4–5 µm thick slices using a microtome and placed on slides. The slides were then dried at 60°C for about 30 minutes to enhance adhesion to the slides. Subsequently, H&E staining was performed. The sections were deparaffinized, hydrated through a series of graded ethanol solutions, stained with hematoxylin for 3–5 minutes to color the nuclei, rinsed with running water, and stained with eosin for 1–2 minutes to color the cytoplasm and other tissue structures. The sections were then dehydrated in ascending ethanol series, cleared, and mounted. After staining, images were observed and captured using an optical microscope.
The pathological injury of lung tissue was scored using the Smith scoring method. Lung edema, alveolar and interstitial inflammation, alveolar and interstitial hemorrhage, atelectasis, and hyaline membrane formation were semi-quantitatively analyzed with a score ranging from 0 to 4; where no injury scored 0, lesion area < 25% scored 1, 25%-50% scored 2, 50%-75% scored 3, and lesion filling the field of view scored 4. The total lung injury score was the sum of these items, with the average score calculated from 10 high-power fields per animal.
Quantitative Real-Time PCR Analysis
The quantitative Real-Time PCR (qRT-PCR) technique was employed to analyze the mRNA expression levels of specific genes. Initially, total RNA was extracted from samples such as cells and tissues using an RNA extraction kit (Takara, cat. no. 9109), following the manufacturer’s guidelines. The concentration and purity of the extracted RNA were determined using a spectrophotometer to ensure RNA quality. Prior to qRT-PCR, the extracted total RNA was transcribed into cDNA using reverse transcriptase and specific primers as per the instructions of the reverse transcription kit (Takara, cat. no. RR047A). Upon completion of reverse transcription, the cDNA was used as a template for qRT-PCR analysis. The qRT-PCR reaction was carried out in a total volume of 10 µL, following the reaction system setup as described in the kit instructions (Takara, cat. no. RR420A). To quantify the expression levels of target genes, the internal reference gene β-actin was used as a control. The expression level changes of the target genes were calculated using the relative quantification method (2^-ΔΔCt method), by comparing the threshold cycle numbers (Ct values) of the target genes with that of the internal reference gene.
Enzyme-Linked Immunosorbent Assay
Enzyme-linked immunosorbent assay (ELISA) technology was utilized for the quantitative analysis of specific protein expression levels. An appropriate ELISA kit was selected based on the target protein. All procedures were conducted strictly following the instructions provided by the kit manufacturer.
Immunohistochemistry and HALO Quantitative Analysis
Immunohistochemistry was employed to localize and quantitatively analyze specific proteins in mouse lung tissues. Following experimental treatment, mice were euthanized, and lung tissues were rapidly excised. The excised tissues were fixed in 4% paraformaldehyde solution for 24 hours, then dehydrated, cleared, and embedded in paraffin. The paraffin-embedded tissues were sectioned into continuous 4–5 µm thick slices and placed on slides. The sections underwent deparaffinization and hydration, followed by antigen retrieval using microwave treatment to expose protein epitopes. Subsequently, the sections were treated with 3% hydrogen peroxide for 10 minutes to block endogenous peroxidase activity. The sections were then blocked with 5% BSA at room temperature for 30 minutes to prevent non-specific binding. Next, the sections were incubated with primary antibodies specific to the target protein, typically overnight at 4°C. The following day, after washing the sections, they were incubated with the corresponding secondary antibodies for 1 hour. Color development was then performed using a diaminobenzidine (DAB) chromogen system, where DAB reacts with hydrogen peroxide to produce a brown precipitate, marking positive signals. Finally, the nuclei were counterstained with hematoxylin and the sections were mounted.
For quantitative analysis of the immunohistochemically stained sections, HALO image analysis software was used. Images of the sections under the microscope were imported into the HALO software, and the software's built-in algorithms were employed to quantitatively analyze the positive signals of the specific protein. This analysis included assessment of the intensity of positive staining, distribution range, and the number of positive cells. By comparing these parameters across different experimental groups, quantitative evaluation of the expression differences of specific proteins in lung tissues was conducted.
ATP Quantitative Analysis in MPMECs
The ATP levels in MPMECs were quantitatively analyzed using an ATP Assay Kit (Elabscience, cat. no. E-BC-F002). Initially, MPMECs were cultured to the required density and subjected to appropriate treatments as per experimental requirements. After treatment, cells were lysed using cell lysis buffer to release ATP. To ensure efficient lysis, the lysed cells were incubated on ice for 10–15 minutes and subjected to repeated pipetting or gentle vortexing for thorough contact with the lysis buffer. Following the instructions of the ATP assay kit, the cell lysate was centrifuged (10000×g, 4°C, for 5 minutes) to remove cell debris. The supernatant was then used for ATP measurement.
In a 96-well plate, 100 µL of sample and ATP detection reagent were added and mixed well, followed by incubation at room temperature for 5–10 minutes. The luminescence intensity was measured using a luminometer, and the ATP concentration in the samples was calculated based on a standard curve. Each sample was assayed in triplicate to ensure the accuracy and reproducibility of the results.
Immunofluorescence Analysis of FAS Protein in MPMECs
Immunofluorescence staining was used to analyze the expression of FAS protein in MPMECs. Initially, MPMECs were cultured on microscope-compatible slides and treated according to experimental groups. Upon reaching appropriate confluency, the cells were fixed with 4% paraformaldehyde solution (Biosharp, cat. no. BL539A) for 20 minutes to preserve cellular morphology and structure. After fixation, cells were washed three times with PBS to remove the fixative. The cells were then permeabilized with 0.1% Triton X-100 (Beyotime, cat. no. P0096) for 10 minutes to allow antibody penetration. Following permeabilization, the cells were again washed with PBS.
To prevent non-specific binding, cells were blocked with 1% BSA (BioFroxx, cat. no. 4240GR100) and incubated for 30 minutes. Subsequently, the cells were incubated overnight at 4°C with a specific primary antibody against FAS protein (Servicebio, cat. no. GB12089-100). The next day, cells were washed with PBS to remove unbound primary antibody, followed by incubation with a fluorescently-labeled secondary antibody (Abcam, cat. no. ab150120) for 1 hour. After secondary antibody incubation, cells were washed again with PBS. Finally, for nuclear staining, DAPI fluorescent dye (Beyotime, cat. no. C1005) was applied. The expression and localization of FAS protein were observed and captured using a fluorescence microscope.
Mouse Lung Wet-to-Dry Weight Ratio
To assess the extent of pulmonary edema, we measured the wet-to-dry weight ratio of the mouse lungs. Initially, mice were subjected to the respective experimental treatments. After the completion of treatments, the mice were euthanized, and their total body weight was accurately measured. The thoracic cavity was then immediately opened to excise the lung tissues. Upon extraction, the lung tissues were first gently rinsed at room temperature with saline to remove blood and adherent materials from the lung surface, followed by gentle dabbing with filter paper to remove surface moisture. Subsequently, the wet weight of the lungs was quickly and accurately measured using a precision electronic balance to ensure accuracy. The lung wet-to-dry weight ratio was calculated by dividing the wet weight of the lungs by the total body weight of the mouse, expressed in mg/g.
Evans Blue Permeability Assay in Mouse Pulmonary Microvasculature
To assess the permeability of the pulmonary microvasculature in mice, an Evans Blue dye assay (Solarbio, cat. no. IE0280) was conducted. Prior to the experiment, mice underwent the designated treatments. At specific time points during the experiment, Evans Blue dye was injected via the tail vein at a dosage of 30 ug/g body weight. After the dye injection, it was allowed to circulate in the system for 30 minutes, ensuring adequate time for the dye to bind to plasma albumin and reach equilibrium. Following the injection period, mice were euthanized, and lung tissues were rapidly excised. The lung tissues were first gently rinsed at room temperature with saline to remove surface blood and adherent materials. Subsequently, 100 mg of minced lung tissue was fixed in 1 mL of methanol for 24 hours to extract Evans Blue dye from the tissue. The absorbance of the solution was then measured at a wavelength of 620 nm using a spectrophotometer to quantify the Evans Blue content in the lung tissues. The content of Evans Blue in the lung tissues was calculated by comparing the measured absorbance values with a known concentration Evans Blue standard curve and expressed in µg/mL wet weight.
Statistical Analysis
Data analysis was conducted using GraphPad Prism (Version 9.0.2). The statistical significance between the experimental and control groups was determined through the use of independent sample t-tests for pairwise comparisons and one-way ANOVA for analyses involving multiple groups. Post-hoc analysis for significant ANOVA results was carried out using Tukey's test to compare means between groups. A threshold of p < 0.05 was established for statistical significance, ensuring that findings were rigorously evaluated for their reliability and validity within the context of our research objectives.