Abstract Mechanical ventilation contributes to diaphragm atrophy and muscle weakness, which is referred to as ventilator-induced diaphragmatic dysfunction (VIDD). The pathogenesis of VIDD has not been fully understood until recently. The aim of this study was to investigate the effects of 24 h of mechanical ventilation on fibro-adipogenic progenitor (FAP) proliferation, endothelial-mesenchymal transition (EndMT), and immune cell infiltration driving diaphragm fibrosis in a rabbit model. The rabbits were anaesthetized and randomly divided into two groups (n = 3 each group): a control group and an experimental group. Diaphragm nuclei for sequencing were prepared by dissociating and filtering muscle tissue. 10X Genomics Platform for single-nucleus RNA sequencing (snRNA-seq) was used to profile the cells. Normalization and clustering were performed by Seurat, and clusters were manually annotated as different cell types. In this study, we performed differentially expressed genes (DEGs) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, pseudotime analysis and high dimensional weighted gene coexpression network analysis (hdWGCNA) to identify the key genes and signaling pathways related to the pathogenesis of VIDD. We further performed quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting to verify the results of snRNA-seq. The snRNA-seq results showed that acute postmechanical ventilation diaphragm cell changes included an increase in the proportion of fibroblasts and a decrease in the proportion of myofibres. The DEGs, KEGG, hdWGCNA and pseudotime analyses demonstrated that fibro-adipogenic progenitor (FAP) proliferation, endothelial-mesenchymal transition (EndMT) and immune cell infiltration are the three main processes involved in early stage of fibrosis development, among which Pdgfd, Sema3a, Cxcr2, are the corresponding regulatory genes. Glycolysis and the gene Pfkfb3 are also important metabolic factors for fibrosis formation. Negr1 and Mef2c are involved in phrenic nerve ending loss and diaphragm fibre atrophy. The qRT-PCR data showed that the mRNA levels of the genes Pdgfd, Cxcr2, Pfkfb3 and Negr1 were significantly greater in the experimental group than in the control group (P < 0.01), and the expression levels of Sema3a and Mef2c were significantly lower (P < 0.01). Despite limitations, including the lack of functional evaluations to confirm ventilator-induced diaphragm dysfunction (VIDD) and the absence of data validating diaphragm unloading during ventilation, our findings suggest that FAP proliferation and immune cell infiltration may play a role in the early stage of driving diaphragm fibrosis during mechanical ventilation. However, future studies are needed to confirm these findings and investigate the potential mechanisms underlying them. Supplementary Information The online version contains supplementary material available at 10.1038/s41598-024-82530-4. Keywords: Single-nucleus RNA sequencing, Diaphragm fibrosis, Fibro-adipogenic progenitor, High dimensional weighted gene coexpression network analysis, Endothelial-mesenchymal transition Subject terms: Molecular medicine, Diagnostic markers Introduction Invasive mechanical ventilation (MV) was commonly used to treat the increased number of patients with acute respiratory distress syndrome during the COVID-19 pandemic in the ICU^[34]1. MV induces diaphragmatic muscle fibre atrophy and profound muscle weakness, termed ventilator-induced diaphragmatic dysfunction (VIDD), which results in delayed weaning and substantial morbidity and mortality. Reactive oxygen species (ROS) activate proteolytic muscle proteases, including caspases and calpains, deregulate the ubiquitin proteasome system and enhance autophagy, which is linked to a decrease in diaphragm protein synthesis and accelerated proteolysis, which are major contributors to VIDD^[35]2. Bruells’s results indicated that diaphragm muscle fibre size increased and contractile function recovered following 12 h of spontaneous breathing as a treatment for VIDD. The recovery of the diaphragm allows the re-establishment of cellular homeostasis of protein synthesis and breakdown^[36]3. In the context of Duchenne muscular dystrophy, fibrosis emerges as a pathological repair process that occurs more frequently than normal diaphragm regeneration, and this fibrosis directly impacts muscle force and physical performance, contributing to the progressive decline in muscle function characteristic of the disease^[37]4. In ventilated critically ill patients with COVID-19, Shi observed for the first time that fibrosis quickly develops in the diaphragm compared with that in patients without COVID-19^[38]5. Qian demonstrated that positive end-expiratory pressure (PEEP) application for 48 h causes collagen deposition and fibrosis in the diaphragm of mechanically ventilated rabbits^[39]6. Shi observed that ECM partially replaces the area lost by myofiber atrophy to provide structural stability of the myofibers and fibrosis quickly develops in the diaphragm of ventilated critically ill patients during ICU stay^[40]7. These findings suggest that diaphragmatic fibrosis is a complex pathological process that can occur in various situations and there are the potential link between VIDD and fibrosis which may lead to diaphragmatic stiffness and decreased contractility. The diaphragm is a complex heterogeneous tissue composed of multinucleated myofibres and mononuclear cells, including immune cells, endothelial cells, muscle stem cells (satellite cells), and nonmyogenic mesenchymal progenitor (e.g., fibro-adipogenic progenitor, or FAP)^[41]8. However, little is known about the mechanism by which the diaphragm recovers from VIDD and the response of cell populations such as satellite cells and FAP to injury. High-throughput spatial transcriptomics of mouse diaphragm cells revealed the activation of Myogenin 1 (Myod1)-expressing satellite cells near the injury site 2 days after injury^[42]9. FAP expressing the markers platelet-derived growth factor receptor alpha (Pdgfrα) and Cd34 are interstitial populations of mesenchymal cells that can differentiate into fibroblasts and adipocytes^[43]10. Fibroblasts are the principal cell types that regulate ECM formation and the process of fibrosis. In response to tissue injury caused by mechanical ventilation, activated FAP proliferate more quickly than satellite cells, which may contribute to diaphragm fibrosis^[44]11. Transcriptome studies on skeletal muscle were conducted using bulk RNA-seq and high-throughput single-cell or snRNA-seq. Bulk RNA-seq provides only averaged gene expression profiles, which inhibits the ability to identify distinct cell types and their dynamics. Single-cell RNA sequencing (scRNA-seq) has been widely utilized to explore different cell types and developmental trajectories based on the diversity of cellular states and to broaden our understanding of cellular heterogeneity in skeletal muscle^[45]12,[46]13. Skeletal muscle is unique in its cellular composition because it is composed primarily of multinucleated myofibres, which are not easily isolated via single-cell dissociation due to their large size and elongated shape. SnRNA-seq has been applied to multinucleated myofibres for transcriptome studies as an alternative to scRNA-seq^[47]14,[48]15. Weighted gene coexpression network analysis (WGCNA) is a biological data analysis tool for constructing gene coexpression networks and identifying core genes that are considered potential disease drivers and are suitable for bulk RNA sequencing data. High-dimensional weighted gene coexpression network analysis (hdWGCNA) is a tool that offers various functionalities for identifying robust modules and hub genes and deciphering their associated biological functions across multiple cell scales and was more suitable to analyse the snRNA-seq data in this study^[49]16,[50]17. The cellular and molecular mechanisms of diaphragm fibrosis and atrophy during mechanical ventilation are unclear. In this study, we profiled the transcriptomic landscape of the diaphragm via snRNA-seq on the 10X Genomics Platform and performed hdWGCNA after one day of mechanical ventilation to determine the major diaphragm cell types, gene regulatory networks and signaling pathways changes. We aimed to evaluate the effects of 24 h of mechanical ventilation on fibro-adipogenic progenitor (FAP) proliferation, endothelial-mesenchymal transition (EndMT), and immune cell infiltration driving diaphragm fibrosis in rabbits. By using snRNA-seq, we hoped to gain insights into the cellular and molecular mechanisms underlying the early changes that occur in the diaphragm during mechanical ventilation. Methods Animals The rabbits were purchased from Shanxi Medical University. All animals weighing between 2.0 and 2.5 kg had ad libitum access to food and water. The rabbits were housed at 22 °C with a humidity of 30–70% under a 12 h light/12 h dark cycle. All animal care and experimental procedures associated with this research were approved by the Institutional Animal Care and Use Committees at Changzhi Medical College, and all experiments were performed in accordance with ARRIVE guidelines. Experimental protocols The rabbits were anaesthetized with pentobarbital sodium (intraperitoneal injection, 50 mg/kg). After induction of anaesthesia, the rabbits were randomly divided into two groups (n = 3 each group): a control group with only anaesthetized spontaneously breathing animals and an experimental group with anaesthetized and mechanically ventilated ones. The ventilator (Harvard Apparatus, Holliston, MA, USA) was used in pressure-controlled mode with a PEEP of 3 cm H[2]O, a respiratory frequency of 40 breaths/min and an inspiratory: expiratory ratio of 1:2. At 12 and 24 h, blood gas analysis was performed for all the animals. The parameters were adjusted to maintain the normal range of PaO[2] and PaCO[2]. An arterial catheter was inserted into the femoral artery to measure arterial blood pressure. Body temperature was maintained at 37 °C using a temperature-controlled heating plate. After 24 h of mechanical ventilation, the animals were euthanized, and diaphragm samples were removed. The tissues were flash-frozen in liquid nitrogen and stored at -80 °C until sufficient nuclei were collected for sequencing. In previous studies we already successfully established a 24-hour mechanical ventilation rabbit diaphragm rest animal model^[51]18. Single-nucleus RNA sequencing We used snRNA-seq to analyse rabbit diaphragm cells from the control (n = 2) and MV (n = 2) groups (OE Biotech Co., Ltd. Shanghai, China). Nuclei were isolated from four samples of dissociated diaphragm tissue (10–15 mg). The cells were placed in lysis buffer containing 0.1% NP40, 10 mM Tris HCl, 146 mM NaCl, 1 mM CaCl[2], 21 mM MgCl[2], and 40 U/mL RNase inhibitor for 7 min. Complete nuclear lysis was confirmed using trypan blue staining, and 1 ml of ST wash buffer was added. The samples were then filtered through a 40 μm cell strainer and centrifuged (500×g for 10 min at 4 °C) to form a crude pellet. The resultant nuclear suspension was pelleted in 100 µl of PBS and 1.0% BSA with a light centrifugation step (250×g for 5 min at 4 °C). Nuclei were counted under a microscope. The cell nuclei suspension was diluted with PBS + 1% BSA to a concentration of 700–1200/µL to reach the optimal range for loading on the 10X Genomics Chromium Chip. The concentration at which nuclei are loaded onto the 10X Genomics Platform is an important parameter affecting the number of nuclei available for downstream analysis. The nuclei were then loaded into the 10X Genomics Chromium Chip using Next GEM Single Cell 3ʹ Kits v3.1 (1000268) to perform cDNA library amplification. cDNA libraries were constructed using a Chromium Next GEM Chip G Single Cell Kit (1000020), and cDNA quality was evaluated by fragment analysis (5200 Fragment Analyser System; Agilent). The constructed library was sequenced using PE150 sequencing mode on an Illumina Nova 6000 platform following the manufacturer’s instructions. Single-nucleus data analysis Illumina sequencing outputs were generated for the samples as binary base call (BCL) files. Using the 10X Genomics Cell Ranger software pipeline (version 3.1.0), the BCL files were demultiplexed and converted into FASTQ files. The raw snRNA-seq FASTQ files were processed to produce a matrix of gene counts versus nuclei. Nuclei expressing more than 200 genes were retained. Moreover, nuclei with more than 5% of the expressed mitochondrial genes were filtered out. Library size normalization was performed on the filtered nuclei using the R package Seurat (version 4.0.0) to obtain the normalized count matrix. Principal component analysis (PCA) was performed to reduce the dimensionality of the log-transformed gene-barcode matrices of the top variable genes. The dimension reduction algorithm mutual nearest neighbours (MNN) was used to eliminate batch effects. Analyses of dimension reduction clustering and cluster-specific marker detection were performed with Seurat. Uniform manifold approximation and projection (UMAP) using the Loupe cell browser (10X Genomics) was used for initial visualization of the optimal cell population through variable gene selection. The generated clusters were identified as specific cell types based on the cell type-specific marker genes, which entailed the canonical markers for individual muscle cell types or the top DEGs in each cluster. The clusters were further classified into subclusters based on the heterogeneity of the specific cell types. DEGs analysis was performed for each of the cell clusters using Seurat. A P value < 0.05 and |log[2]foldchange|>0.58 were set as the thresholds for significant differential expression. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of the DEGs was considered significant at an adjusted P value < 0.05. DEGs and KEGG analyses revealed key candidate genes and molecular pathways in the regulatory network related to diaphragm fibrosis and atrophy. To infer dynamic gene expression and progression among the clusters and subclusters, we utilized the R package Monocle (version 2.9.0) to perform pseudotime trajectory analysis. We used the differential gene test function of the Monocle package to select genes (q < 0.01) that were likely to be informative in the ordering of cells along the pseudotime trajectory. We then applied protein‒protein interaction (PPI) analysis to the hub and differential genes, which indicated that genes situated at the center of the interaction network were responsible for their key connections with other genes. High-dimensional WGCNA To investigate the intrinsic properties of the snRNA-seq data, hdWGCNA was performed to decipher the hub genes expression. Following the workflow for conducting hdWGCNA, we constructed gene coexpression networks based on pairwise correlations among genes using snRNA-seq data from four diaphragm samples. The WGCNA tree has a minimum size of 50 genes, and similar genes are integrated into one module. Modules or clusters of highly correlated genes were identified, and the module eigengenes (MEs) describing the expression patterns of coexpression modules were calculated. The genes with the highest eigengene-based connectivity (kME) values in the modules were identified as hub genes. Quantitative real-time polymerase chain reaction (qRT-PCR) To confirm the results of the snRNA-seq analysis, the expression of six genes, 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (Pfkfb3), Platelet-derived growth factor D (Pdgfd), C-X-C Motif Chemokine Receptor 2 (Cxcr2), Neuronal growth regulator 1(Negr1), Semaphorin-3a (Sema3a) and myocyte enhancer factor-2 C (Mef2c), were determined using qRT-PCR. TRIzol reagent (15596026, Ambion, Carlsbad, CA, USA) was used for total RNA extraction from whole-diaphragm samples according to standard protocols. The quality of the purified RNA samples was evaluated with an Agilent Bioanalyzer (Agilent Technologies, Inc., Santa Clara, CA, USA), and RNA samples with an RNA integrity number (RIN) > 8.0 were used for gene expression measurements. Reverse transcription was performed with the Super Script II First-Strand Synthesis Kit (2690A, Takara Bio USA Inc., San Jose, CA, USA), and qRT-PCR was performed with the SYBR Fast Universal qPCR Kit (KK4601, KAPA Biosystems, Woburn, MA, USA) on a CFX-Connect 96 Real-Time PCR detection System (Bio-Rad, Hercules, CA, USA). The primer sequences (5’-3’) used were as follows: Pfkfb3, forward: AAACGCCTTAAGCTCCTAGTGAT, reverse: GCAACGTGCCAGTCTGATGG;Pdgfd,forward: ACCTGGGTTCAAAAGAATTTA, reverse: GACTTGCGATCATGGTAT;Cxcr2,forward: CAACTCCCTGGTGATGCT, reverse: TCCTTCACAAGCGAGACC;Negr1,forward: ACCAGAGCCTTCCATTTC, reverse: GGTCACAGTGCCAGATTTA;Sema3a,forward: GATGAGGAACGGAGTAGGC, reverse: GCAAATTGGGTGGAAAGC;Mef2c,forward: CCCAACCTCTTGCCACTG, reverse: TTGCCATATCCGTTCCCT. The relative gene expression was quantified by the ΔΔCt method, with the gene Gapdh serving as the internal control. Western blotting analysis A maximum of 20 mg of diaphragm tissue was homogenized on ice in 800 ul of lysis buffer. Total diaphragm proteins were isolated using RIPA buffer (R0010, Solarbio, Science & Technology Co., Ltd., Beijing, China) supplemented with protease inhibitor cocktails. After centrifugation (12,000 rpm for 15 min), the protein concentration was determined using a bicinchoninic acid (BCA) protein assay kit (P0010, Solarbio). Subsequently, 20 µg of each sample was subjected to 10% SDS‒PAGE and transferred to a PVDF membrane (IPVH00010, Millipore, Billerica, MA). After the semidry blotting procedure (50 min, 90 V), the membrane was incubated for 1 h at room temperature (RT) in 5% BSA blocking solution, followed by overnight incubation on a shaker at 4 °C with primary antibodies against PFKFB3 (bs-3528R, Boster Biological Technology Co., Ltd., Wuhan, China), PDGFD (bs-24572R, Boster), CXCR2 (bs-1629R, Boster), NEGR1 (bs-11095R, Boster), SEMA3A (bs-10468R, Boster), MEF2C (bs-4130R, Boster) and β-actin (66009-1-Ig, Proteintech, Wuhan, China) as a loading control. The membrane was then incubated for 1 h at room temperature on a shaker with a horseradish peroxidase-conjugated goat anti-rabbit secondary antibody (BL003A; Biosharp, Guangzhou, China). After the bands were visualized with tetramethylbenzidine, the blottings were scanned and quantified using Tanon GIS software (version 4.2; Tanon Technology, Shanghai, China). Statistical analysis Statistical analysis was performed with SPSS (version 25.0; SPSS Inc., Chicago, IL, USA). Differences between two groups were evaluated using two-tailed Student’s t-tests. All results are presented as the mean ± standard deviation (SD). A P value < 0.05 was considered statistical significance. Results Blood gas and arterial blood pressure Blood gas analysis revealed that the PaO[2] concentration remained at 133 ± 11 mmHg for the experimental group and 125 ± 12 mmHg for the control group. This was also the case for the PaCO[2] concentration (experimental group, 35 ± 6 mmHg; control group, 33 ± 5 mmHg) and pH (experimental group, 7.44 ± 0.03; control group, 7.41 ± 0.02). Arterial blood pressure was similar in the different groups and averaged 115 ± 4 and 127 ± 6 mmHg for the experimental group and control group, respectively. Diaphragm cell clusters and subclusters Our snRNA-seq data revealed the intricate transcriptional dynamics of the diaphragm during 24 h of mechanical ventilation. Nuclei from rabbit diaphragms were purified and successfully constructed on the 10X Genomics Platform (Fig. [52]1). We profiled 14,652 nuclei transcriptomes from the diaphragms of 2 mechanically ventilated rabbits and 2 controls for analysis following normalization and quality control of the snRNA-seq data. Sequencing data were plotted using UMAP to generate functionally enriched clusters. Subsequently, the generated clusters were annotated according to the expression of cell type-specific marker genes. The fibre type-specific marker genes were identified as previously reported or by the top cell type-specific differential genes. We identified 15 clusters annotated to 10 major cell types in the diaphragm (Fig. [53]2a). Two top marker genes for each cluster were displayed on the dot plots (Fig. [54]2b). Violin plots showed that the marker genes Pfkfb3 and Tbc1d1 were mainly expressed on the clusters 1,2,8, Tek and Emcn on the clusters 9,14 (Fig. [55]2c). Clusters 1, 2, 8 were annotated as fibroblast 1 with the marker genes Pfkfb3 and Tre-2/USP6, BUB2, cdc16 domain family member 1 (Tbc1d1) (Fig. [56]2d); Cluster 5 was defined as myocyte 1 with the marker genes Myh7b and Mylk3 (Fig. [57]2e); Cluster 7 was defined as myocyte 2 with the marker genes Myh1 and Myl1 (Fig. [58]2f); Cluster 6 was defined as fibroblast 2 containing FAP with the marker genes Col15a1 and Pdgfd (Fig. [59]2g); Clusters 9 and 14 were defined as endothelial cells marked by Tek and Emcn (Fig. [60]2h), Cluster 9 has a much more distinct expression profile and was defined as postcapillary venule (PCV) endothelial cells; Cluster 12 was defined as neurocytes marked by Pak5 and Ntng1 (Fig. [61]2i); Cluster 13 was divided into immune cells marked by Ikzf1 and Cd163 (Fig. [62]2j) and satellite cells marked by Pax7 and Fgfr4 (Fig. [63]2k); Cluster 15 was annotated as pericytes and smooth muscle cells marked by Acta2 and Rgs5 (Fig. [64]2l). We further divided the marked cell population into cell subgroups. The biomarkers included global markers, which specify cell types, and local markers, which specify subtypes. The diaphragm has three different specialized fibroblast clusters: mature fibroblasts that are shared by all tissue fibroblasts in the steady state, intermediate fibroblasts between activated and quiescent states, and naive fibroblasts that are activated by injury. The cell population of fibroblasts that increased within 24 h of mechanical ventilation was named fibroblast 1, which consisted of two subclusters: myofibroblasts-myoblasts and naive fibroblasts (Fig. [65]3a, b, c). Endothelial cells can be separated into two subclusters: main and PCV endothelial cells (Fig. [66]3d, e, f). The clusters with high Pdgfd expression were identified as fibroblast 2, which included two subclusters: mature and intermediate fibroblasts (Fig. [67]3g, h, i), FAP were included in fibroblast 2. The small populations of immune cells and satellite cells in the samples were separated into immune cell subclusters and satellite cell subclusters (Fig. [68]3j, k, l). Fig. 1. [69]Fig. 1 [70]Open in a new tab Schematic diagram for single-nucleus sequencing and diaphragm fibrosis formation. a Schematic diagram for nuclei purification and sequencing from rabbit diaphragms (By Figdraw 2.0, ID: AORYU13345). b Clustering of snRNA-seq data through uniform manifold approximation and projection (UMAP) dimension reduction analysis from a combined set of four samples. 15 clusters annotated into 10 cell types, namely, fibroblast 1, fibroblast 2, myocyte 1, myocyte 2, endothelial cells, neurocyte, immune cell and satellite cell, pericytes and smooth muscle cell. Cells are coloured by major cell type assignment. The proportion of fibroblast 1 cells increased, and the proportion of myocytes decreased quickly after 24 h of mechanical ventilation in the diaphragm. c Schematic diagram of diaphragm fibrosis during 24 h of mechanical ventilation (By Figdraw 2.0, ID: ITAOR9fdde). d Schematic diagram showing the key gene signatures and signaling pathways associated with the development of diaphragm fibrosis and atrophy (By Figdraw 2.0, ID: WTAOWa775f). Fig. 2. [71]Fig. 2 [72]Open in a new tab Diaphragm cell type identification. a Clustering algorithm with a resolution of 0.6 identified 15 clusters. b Dot plots of key marker gene expression levels across cell clusters. The average expression (dot colour intensity) and cell percentage (dot size) of the two top marker genes for each cluster were compared. c. Violin plots showing the relative expression levels of selected genes for different clusters. Cells are coloured according to the expression of the specific marker genes. UMAP of snRNA-seq data displayed the presence of Fibroblast 1 containing Cluster 1, 2, 8 marked by Pfkfb3 and Tbc1d1 (d); Myocyte 1 corresponding to Cluster 5 marked by Myh7b and Mylk3 (e); Myocyte 2 corresponding to Cluster 7 marked by Myh1 and Myl1 (f); Fibroblast 2 corresponding to Cluster 6 marked by Col15a1 and Pdgfrα (g); Endothelial cell including Cluster 9,14 marked by Tek and Emcn (h); Neurocyte corresponding to Cluster 12 marked by Pak5 and Ntng1 (i); Immune cell corresponding to Cluster 13 marked by Ikzf1 and Cd163 (j); Satellite cell corresponding to Cluster 13 marked by Pax7 and Fgfr4 (k); Pericytes and smooth muscle cell corresponding to Cluster 15 marked by Acta2 and Rgs5 (l). Fig. 3. [73]Fig. 3 [74]Open in a new tab Subcluster and cell type analysis. Clustering and identification of subpopulations. Specific cell populations, such as Fibroblast 1 (a), Endothelial cell (d), Fibroblast 2 (g), Immune and Satellite cell (j), were marked with Loupe software, while other cell populations are marked in grey. Fibroblast 1 has two main subclusters (b): Myofibroblasts/myoblasts, which are marked by the genes Tns1 and Myod1, and naive fibroblasts, which are marked by Sec24d and Fbxo32 (c). Endothelial cells also had two main subclusters (e): endothelial cells marked by the Adamts12 and Foxp2 genes and PCV endothelial cells marked by Dach1 and Mecom (f). Fibroblast 2 has two main subclusters (h): intermediate fibroblasts, which are marked by the genes Pdk4 and Zbtb16, and mature fibroblasts, which are marked by Dtx3 and Phldb2 (i). There are two main subclusters of immune and satellite cells (k): satellite cells, marked by the genes Fgfr4 and Pax7, and immune cells, marked by Ikzf1 and Cd163(l). DEGs and KEGG analysis We performed DEGs and KEGG pathway enrichment analysis (Fig. [75]4). As shown in the volcano plots, the expression of Pfkfb3, Tbc1d1, Pdgfd, and Cxcr2 was significantly upregulated in the experimental group compared with the control group in the fibroblast 1, fibroblast 2 and immune cells (P < 0.05), while the expression of Ccl21, Mef2c, and Negr1 was significantly downregulated in the experimental group compared with the control group in the endothelial cells, myocyte and neurocyte respectively (P < 0.05). KEGG analysis revealed that the DEGs in the fibroblast 1, fibroblast 2, immune cell, endothelial cell, myocyte and neurocyte were functionally enriched in the insulin signaling pathway, phosphatidylinositol 3-kinase (PI3K) -Akt signaling pathway, phosphalipase D (PLD) and Rap1 signaling pathway, chemical carcinogenesis-reactive oxygen species, calcium signaling pathway and leukocyte transendothelial migration. Fig. 4. [76]Fig. 4 [77]Open in a new tab Differentially expressed genes and Kyoto Encyclopedia of Genes and Genomes analysis. Volcano plots showing the top twenty up- or downregulated genes for each cell type. The red dots represent upregulated genes, and the blue dots represent downregulated genes. (P value < 0.05, and |log2foldchange| > 0.58). Bubble plot indicating the top enriched pathways for each cell type based on KEGGpathway enrichment analysis of differentially expressed genes([78]www.kegg.jp/kegg/kegg1.html). The sizes of the dots represent the number of genes included in each pathway. The colour gradient of dots represents the adjusted P values of each enriched pathway. The genes Pfkfb3, Tbc1d1 and the insulin signaling pathway; Pdgfd and the PI3K-Akt signaling pathway; Cxcr2 and PLD, Rap1 signaling pathways; Ccl21 and chemical carcinogenesis-reactive oxygen species signaling pathway; Mef2c and the calcium signaling pathway; Negr1 and leukocyte transendothelial migration signaling pathway are labelled by the black and red bars in Panels a, b, c, d, e, and f. hdWGCNA and PPI network We performed hdWGCNA on a single-nucleus transcriptomic dataset of diaphragm tissue from two mechanically ventilated rabbits and two control ones, which revealed distinct network structures and sets of gene modules. After weighting the median connectivity and scale-free topology model fit, a power value of 6 was selected. The final analysis resulted in eight modules that were derived based on a scale free net-work structure. Within cell type-specific modules, the hub genes exhibited the highest intramodular connectivity among the coexpression networks and displayed biologically relevant information for each major cell type. We identified brown module was consistent with the fibroblast 1 cluster, containing two labelled hub genes, Pfkfb3 and Tbc1d1; the green module was consistent with the fibroblast 2 cluster, containing a labelled hub gene, Pdgfd; the turquoise module was consistent with the myofiber cluster, containing a labelled hub gene, Ryr3; and the yellow module was consistent with the endothelial cell cluster, containing a labelled hub gene, Sema3a (Fig. [79]5). Our analysis ultimately led to the identification of five hub genes: Pfkfb3, Tbc1d1, Pdgfd, Ryr3, and Sema3a. We constructed nine PPI networks centred on the proteins PFKFB3, TBC1D1, PDGFD, CXCR2, SEMA3A, CCL21, NEGR1, MEF2C, and RYR3 to determine their relationships with other proteins (Fig. [80]6). Fig. 5. [81]Fig. 5 [82]Open in a new tab High-dimensional weighted gene coexpression network analysis (hdWGCNA). a The top left panel depicts the soft power threshold for choosing a scale-free topology model. The average connectivity of the topological network was most stable at the lowest soft threshold of 6. b Dynamic Tree Cut algorithm for gene clustering. Each leaf on the tree represents a gene, and the colour at the bottom indicates the assignment to a specific coexpression module. c Bubble plots displaying the scores obtained for eight modules in eight cell subtypes. d snRNA-seq UMAP coloured by module eigengene (ME) for eight coexpression modules. e The eight modules’ values of -log10 (Adj P value) on the Y-axis versus average log2 (fold change) on the X-axis are shown for each cell type, including endothelial cells, fibroblast 1, fibroblast 2, myocytes, neurocytes, pericytes, satellite cells and smooth muscle cells. The larger the value is, the greater the fit between the coloured module and the specific cell type. The plot shows that fibroblast 1 features a brown module, fibroblast 2 features a green module, endothelial cells feature a yellow module, and myocyte cells feature a turquoise module. f UMAP plot of the gene coexpression network with eight coloured modules and the top two hub genes labelled in each module. Nodes are coloured according to coexpression module assignment. g Hub genes in each module were identified by eigengene-based connectivity (KME). The following five hub genes associated with fibrosis were identified: Pfkfb3 and Tbc1d1 in the brown module, Pdgfd in the green module, Sema3a in the yellow module and Ryr3 in the turquoise module. Fig. 6. [83]Fig. 6 [84]Open in a new tab Protein-protein interaction network (PPI network).We constructed nine PPI networks displaying protein-protein interactions among the related genes. The nodes represent proteins, and the edges represent the interaction strength between two proteins. The proteins PFKFB3 (a), PDGFD (b), CXCR2 (c), CCL21 (d), SEMA3A (e), RYR3 (f), MEF2C (g), NEGR1(h) and TBC1D1 (i) which are located at the hub of the interaction network, are responsible for diaphragm fibrosis and atrophy. Pseudotime trajectory analysis We identified four trajectories corresponding to distinct cell fates, termed the satellite cell, FAP cell, endothelial cell and immune cell trajectories. Pseudotime analysis revealed that the satellite cell trajectory started from Cluster 13 to Cluster 1, indicating that the quiescent satellite cells expressing Pax7 differentiated from progenitors to myoblasts expressed Myod1 after activation during mechanical ventilation. Monocle analysis clarified the satellite cell trajectory by reconstructing two main branches. One trajectory identified cells progressing towards myofiber differentiation, as indicated by the activation of Myod1. Another branch continued to proliferate, retaining the identity of the satellite cell (Fig. [85]7a, b, c). The ordering of cells in the trajectory tree suggested that the FAP cell trajectory seemed to start principally from Cluster 12, then move towards Cluster 11, and finally to Clusters 1, 2, 8 representing the trajectory from FAP cell (contained in fibroblast 2) to naive fibroblasts (contained in fibroblast 1) (Fig. [86]7d, e, f). The trajectory of endothelial cells started principally from Clusters 9 and 14, then moved towards Cluster 10, and finally to Clusters 1, 2, 8 representing the trajectory from endothelial cells to naive fibroblasts (Fig. [87]7g, h, i). The immune cell trajectory started principally from Cluster 13 and then moved to Clusters 1, 2, 8 representing the trajectory from immune cells to naive fibroblasts (Fig. [88]7j, k, l). Fig. 7. [89]Fig. 7 [90]Open in a new tab Single-nucleus trajectory analysis. Pseudotime trajectories reconstructed by Monocle 2 highlight the utility of nucleus-level resolution, which reflects the transcriptional dynamics that regulate the formation of diaphragm fibrosis. a UMAP-coloured only the satellite cell and myoblast; b the trajectory of satellite cells to myoblasts; d UMAP-coloured fibroblast 2, other cells and fibroblast 1; e The trajectory of FAP (contained in fibroblast 2) to naive fibroblasts (contained in fibroblast 1); g UMAP-coloured endothelial cells, other cells and fibroblast 1; h The trajectory of endothelial cells to naive fibroblasts (contained in fibroblast 1); j UMAP-coloured immune cells and fibroblast 1; k The trajectory of immune cells to naive fibroblasts (contained in fibroblast 1). The beginning and end of the timeline in pseudotime trajectories are displayed with the direction of the arrows (c, f, i, l). Quantitative real-time PCR and Western blotting analysis The qRT-PCR data showed that the mRNA levels of the Pfkfb3, Pdgfd, Cxcr2, and Negr1 genes were significantly higher in the mechanically ventilated group than in the control group (P < 0.01), and the expression levels of Sema3a and Mef2c were significantly lower (P < 0.01). Western blotting analysis revealed that the protein expression of PFKFB3, PDGFD, and CXCR2 tended to increase and that the protein expression of SEMA3A, NEGR1 and MEF2C tended to decrease in the experimental group compared with the control group, but these differences did not reach statistical significance (Fig. [91]8). Fig. 8. [92]Fig. 8 [93]Open in a new tab Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. qRT-PCR and Western blotting showing the expression of selected genes and corresponding proteins in the mechanically ventilated diaphragm and control ones. The bar graphs show the quantification of the mRNA expression of Pfkfb3 (a), Pdgfd (b), Cxcr2 (c), Negr1 (d), Sema3a (e), and Mef2c (f) normalized to that of GAPDH. Asterisks indicate significant differences (n = 3 in each group) (*P < 0.05). Western blotting analysis of the protein expression of PFKFB3 (g), PDGFD (h), CXCR2 (i), NEGR1 (j), SEMA3A (k), and MEF2C (l) normalized to that of β-actin as a loading control. Discussion This study is the first attempt to gain insights into the cellular and molecular mechanisms underlying the early changes that occur in the diaphragm in response to mechanical ventilation by snRNA-seq using a rabbit model. We found that after 24 h of mechanical ventilation diaphragm cell changes included a decreased proportion of myofibres and an increased proportion of fibroblasts, which may contributed to the development of diaphragm fibrosis. In contrast to the traditional perspective, the study found evidence of myocyte atrophy and fibrosis in the diaphragm, as well as potential reductions in diaphragmatic contractile force. These findings suggest that myocyte atrophy and fibrosis may jointly contribute to VIDD, but additional studies are needed to confirm their causality and to investigate other potential factors involved in this condition. During 24 h of mechanical ventilation in rabbits, the activation of fibroblasts and their subsequent differentiation into myofibroblasts, which depends on a combination of specific factors induced the development of fibrosis. The genes Pfkfb3 and Tbc1d1 and the insulin signaling pathway may drive glycolysis to meet the energy requirements for fibroblast proliferation during mechanical ventilation. In addition, proliferation of FAP and fibroblast-to-myofibroblast transdifferentiation (FMT) associated with the corresponding genes Pdgfd and the PI3K-Akt signaling pathway, EndMT associated with the corresponding gene Sema3a and the signaling pathway chemical carcinogenesis-reactive oxygen species, immune cell infiltration associated with the regulatory gene Cxcr2, PLD and Rap1 signaling pathways are the three main processes involved in early stage of driving diaphragm fibrosis. Phrenic nerve ending loss resulting in a lack of trophic support with decreased Negr1 gene expression and leukocyte transendothelial migration signaling pathway activation, vascular reduction leading to diaphragm hypoperfusion with decreased Ccl21 gene expression, calcium signaling pathway activation with decreased Mef2c and Ryr3 genes expression are the three main factors responsible for myocyte atrophy. Muscle satellite cells are progenitors of muscle fibres and reside under the basal lamina in direct contact with myofibers^[94]19. In rabbits, the diaphragm muscle shows earlier and more severe fibre damage during mechanical ventilation. Upon injury, quiescent satellite cells began to proliferate and differentiate into myofibroblasts-myoblasts with upregulated Myod1 expression, which was confirmed by the satellite cell trajectory analysis in our study. However, relatively few myogenic precursors expressing Myod1 exhibited a limited proliferation and differentiation capacity to meet functional demands as diaphragmatic fibres are lost. Under these conditions, normal repair of the diaphragm is replaced by fibrosis, which is a pathological repair process. Aerobic glycolysis has been increasingly recognized as an important pathogenic process that contributes to the development of many types of fibrosis^[95]20. Metabolic reprogramming of glycolysis is necessary to meet the energetic demand of activated fibroblasts compared to quiescent fibroblasts, and this metabolic switch highlights the importance of glycolysis for fibrosis development^[96]21. Pfkfb3 is a critical rate-limiting enzyme that promotes glycolysis metabolism by modulating and maintaining the intracellular concentration of fructose-2,6-bisphosphate (F-2,6-BP)^[97]22. Upregulation of Pfkfb3 increased glycolysis through the TGF-β signaling pathway, and the production of lactate directly contributed to fibroblast activation and proliferation in renal fibrosis models of unilateral ureter obstruction and kidney ischaemia/reperfusion^[98]23. The overexpression of Pfkfb3, which promotes glycolysis in cardiac fibroblasts, results in cardiac fibrosis while postmyocardial infarction^[99]24. In an experimental model of pulmonary fibrosis, glycolytic reprogramming promoted lung myofibroblast activation by upregulating the expression of the critical glycolytic enzyme Pfkfb3^[100]25. Tbc1d1 is a member of the TBC1 Rab-GAP family of proteins and has distinctive roles in regulating both insulin- and contraction-stimulated glucose transport in skeletal muscle^[101]26. Insulin and exercise stimulate the phosphorylation of Tbc1d1 by upregulating insulin receptor substrate (IRS)-PI3K and increasing AMP-activated protein kinase activity, and Tbc1d1 phosphorylation facilitates the translocation of glucose transporter 4 (GLUT4) to the cell surface membrane, resulting in increased cellular glucose uptake^[102]27. Pfkfb3 and Tbc1d1 are the critical nodes involved in the insulin signaling pathway and are central regulators of metabolic function in skeletal muscle. The increased expression of Pfkfb3 and Tbc1d1 in our snRNA-seq data suggested that there were strong connections between the transcriptomic and metabolomic profiles of the diaphragm in mechanically ventilated rabbits for 24 h. We speculated that enhanced glucose metabolism driven by Pfkfb3, Tbc1d1 and the insulin signaling pathway may produce a large amount of energy to meet the demand of rapidly differentiating fibroblasts, which is necessary for the development of diaphragm fibrosis. FAP are nonmyogenic mesenchymal stem cells that reside in the interstitial space between myofibres and are characterized by the expression of Pdgfrα, which induces FMT and increases the production of ECM proteins for muscle structure support^[103]28,[104]29. Pdgfd strongly activated the PI3K-AKT pathway. AKT-catalysed phosphorylation of the forkhead box gene, group O1 (Foxo1), results in its translocation from the nucleus to the cytosol and prevents Foxo1-dependent gene expression of manganese superoxide dismutase (Mnsod). The downregulation of Mnsod leads to the excess production of ROS and FMT^[105]30. Previous studies reported that FAP, which are the predominant contributors to pathological diaphragm fibrosis, expressed greater levels of the Pdgfrα protein in the diaphragm than in the limb muscles of mdx mice (a muscular dystrophy model)^[106]31. In the present study, increased Pdgfd expression, as determined by DEGs analysis of single-nucleus sequencing data, was confirmed by qRT-PCR and Western blotting. We postulated that increasing Pdgfd expression together with PI3K/AKT/FoxO1 pathway activation were the main drivers of diaphragm fibrosis in response to muscle injury during mechanical ventilation based on the DEGs analysis via snRNA-seq. Mechanical ventilation triggered the recruitment of circulating immune cells into diaphragmatic tissue after diaphragm injury. In our study, the increase in the expression of Cxcr2 and activation of the Rap1 and PLD signaling pathways could clarify the molecular mechanisms by which immune cells infiltrate diaphragmatic tissue and transform into fibroblasts. Cxcr2 belongs to the γ subfamily of GTP binding proteins (G-proteins) and is the receptor for CXCL8. Once the chemokine CXCL8 binds to the receptor Cxcr2, the β and γ subunits of the G-protein are released and the downstream small GTPase RAS-related protein 1 (Rap1) is activated, PLD is also activated to produce the second messenger phosphatidic acid (PA), which increases immune cell adhesion and migration, this two processes are closely related to the recruitment of immune cells in response to acute diaphragm injury^[107]32,[108]33. Vascular endothelial cells form a monolayer lining the vasculature throughout the body and play diverse roles in maintaining internal homeostasis. EndMT is a transcriptional program that downregulates the expression of endothelial cell marker genes and upregulates the expression of mesenchymal cell marker genes. Endothelial cells can transdifferentiate into fibroblasts via EndMT and contribute to extracellular matrix deposition by driving fibrogenesis^[109]34. SEMA3A is a special membrane-associated secreted protein with various biological properties, and the SEMA3A/LIMK/p-cofilin/actin axis is involved in EndMT. Endothelial cells acquire migratory capabilities after actin remodelling during EndMT. SEMA3A blocks TGF-β-induced EndMT by inhibiting cytoskeletal actin remodelling and cell migration mediated by LIMK/p-cofilin signaling^[110]35. In our study, reduced expression of Sema3a/SEMA3A, as determined by qRT-PCR and Western blotting, increased LIMK/p-cofilin signaling and contributed to TGF-β-induced EndMT via dynamic actin remodelling, thus contributing to the development of diaphragm fibrosis. Taken together, these findings indicate that FAP are the most important contributors to mechanical ventilator-triggered fibroblast transformation and activation as early as 24 h. The contribution of immune cells and endothelial cells to diaphragm fibrosis is likely limited due to the rarity of immune cells, and EndMT is a complex phenotypic transformation process. Negr1 is a glycosylphosphatidylinositol (GPI)-anchored membrane synaptic adhesion molecule highly expressed in the cerebral cortex and hippocampus^[111]36. Negr1 promotes sympathetic axonal growth and affects adult neurogenesis by regulating leukocyte inhibitory factor receptor (LIFR) signaling^[112]37. In addition, leukocyte transendothelial migration (LTM) is an inflammatory process in which leukocytes cross through the blood vessel wall, leading to tissue damage and organ failure^[113]38. LTM induces the apoptosis of neurons by secreting various cytotoxic molecules, such as C-X-C motif chemokine ligand 10 (Cxcl10)^[114]39. In our study, reduced secretion of Negr1 combined with LTM activation resulted in the progressive loss of phrenic nerve terminals through inflammatory cell recruitment and apoptosis promotion during 24 h of mechanical ventilation in the diaphragm. The degeneration of phrenic neuron endings contributes to a lack of trophic support for muscle tissue and diaphragm dysfunction. Angiogenesis refers to the formation of new blood vessels that depend on endothelial cell activation and proliferation via the release of endothelial growth factors^[115]40. Angiogenesis plays a key role in wound healing during the inflammation phase by facilitating the recruitment of inflammatory cells and the secretion of growth factors^[116]41. Ccl21 is the main chemokine that promotes angiogenesis by enhancing the formation of blood vessels through binding to CCR7 and activating the PI3K pathway^[117]42. In our study, Ccl21 expression was reduced at the 24-hour time point in the experimental group compared to the control group. Correspondingly, we propose that reduced Ccl21 expression results in blood vessel loss, especially blood capillary loss, and decreased blood flow leading to diaphragm dysfunction within 24 h of mechanical ventilation. Previous animal studies revealed that slow and fast-twitch diaphragm fibres exhibit atrophy simultaneously induced by MV^[118]43, while our study revealed that slow fibre atrophy is induced by MV via the calcium signaling pathway and the genes Ryr3 and Mef2c. Defective intracellular Ca^2+ handling is central to depressed diaphragm contractility in mechanically ventilated rabbits^[119]44,[120]45. There are three different isoforms of the ryanodine receptor Ca^2+ release channel (Ryr1, Ryr2, and Ryr3) expressed on the sarcoplasmic reticulum in skeletal muscle, and the Ryr1 isoform represents the main isoform of the Ca^2+ release channel^[121]46. In VIDD, diaphragmatic mitochondrial oxidative stress causes Ryr1 channel complex instability and Ca^2+ leakage, then cytoplasmic Ca^2+ overload leads to calpain activation and endoplasmic reticulum (ER) stress, which contribute to myocyte proteolysis and apoptosis^[122]47,[123]48. Ryr3 expression is found only in the slow fibres of the diaphragm in adult rabbits^[124]49. One study showed that RyR1 and RyR3 coexpressed in the same cell in rabbit diaphragm forms a homotetramer^[125]50. The coexpression of the RyR1 and RyR3 calcium release channels in skeletal muscles may result in a diversification of the excitation-contraction coupling apparatus to obtain a fine regulation of muscle contraction^[126]51. In our study, the identification of the turquoise module, which includes the hub gene Ryr3 is a novel finding, given the relatively low expression levels of RyR3 compared to RyR1 in the diaphragm. We propose RyR3 might play the important role leading to diaphragm slow-twitch fibre atrophy and contractile dysfunction in the diaphragm. The myocyte enhancer factor-2 (Mef2) family is a family of transcription factors that regulate skeletal muscle repair and regeneration through the activation of satellite cells. Motor nerve activity is mediated by calcineurin-dependent genes via Mef2 transcription to control fundamental cellular processes, thereby establishing special physiological myofiber subtypes^[127]52. In vertebrates, this family has four members: Mef2a, Mef2b, Mef2c and Mef2d. Mef2c, an essential transcription factor, regulates the transcription of slow-twitch fibre specific genes, particularly Myh7, and its product MyHC-β/slow^[128]53. Prolonged bed rest in humans induces robust inhibition of the Mef2 family members, particularly Mef2c, which ultimately reduces skeletal muscle mass^[129]54. In our study, phrenic nerve ending loss led to a decrease in nerve impulses combined with mechanical ventilator unloading in the diaphragm, which induced Mef2c downregulation and slow-twitch fibre atrophy. In contrast to the traditional perspective, we speculated that diaphragm atrophy may originated from three main important factors, namely, phrenic nerve ending loss and trophic support deletion with the corresponding gene Negr1, diaphragm blood vessel reduction and hypoperfusion with the corresponding gene Ccl21, and calcium signaling pathway activation with decreased Mef2c and Ryr3 expression. Limitations There are several limitations in our study. First, the data emphasized that mechanical ventilation contributed to acute diaphragm fibrosis as early as after 24 h. However, fibrosis is a chronic lethal disorder and requires long-term progression. The extension of research time may help identify additional information that underlies fibrosis progression. Second, snRNA-seq in multinucleated cells is complex because we cannot determine which nuclei originate from the same cell. Additionally, isolating the nucleus means that only a fraction of the transcripts within a cell are surveyed; nuclear resident transcripts such as long noncoding RNAs (lncRNAs) are not included in this type of analysis. Third, there were opposite trends in Negr1 mRNA and NEGR1 protein expression. We speculated that the increase in Negr1 mRNA expression may reflect a transient response followed by decreased expression due to posttranslational modifications. Finally, we did not assess diaphragm muscle fiber injury, atrophy, or diaphragm strength. Therefore, our findings can only be correlated with 24-hour mechanical ventilation in rabbits and not directly with VIDD, atrophy, or injury. Conclusions Our study found that 24 h of mechanical ventilation in rabbits led to changes in FAP proliferation, EndMT, and immune cell infiltration in the diaphragm. However, it is important to note that our findings are speculative and cannot be definitively linked to VIDD due to the limitations of our study. Specifically, we did not perform functional evaluations to confirm the presence of VIDD, nor did we have data to validate diaphragm unloading during the ventilation period. Despite these limitations, our findings suggest that FAP proliferation and immune cell infiltration may play a role in the early stage of driving diaphragm fibrosis during mechanical ventilation. Future studies are needed to investigate the potential mechanisms underlying these changes and their potential contribution to VIDD. Electronic supplementary material Below is the link to the electronic supplementary material. [130]Supplementary Information 1.^ (81.8KB, jpg) [131]Supplementary Information 2.^ (56.6KB, jpg) [132]Supplementary Information 3.^ (107.1KB, jpg) [133]Supplementary Information 4.^ (72.8KB, jpg) [134]Supplementary Information 5.^ (78.7KB, jpg) [135]Supplementary Information 6.^ (77.6KB, jpg) Acknowledgements