1. Introduction
Chronic lung diseases have emerged as the third leading cause of mortality worldwide, with advancing age serving as a significant predisposing factor in the pathogenesis and progression of diverse pulmonary ailments [
1,
2]. Empirical evidence derived from epidemiological inquiries reveals an overwhelming predominance of pneumonia-related deaths among elderly individuals aged 65 years and older, accounting for approximately 85% of such cases. In contrast, the younger demographic below 45 years contributes a mere 3% [
3]. Furthermore, research indicates that individuals aged 80 years and older infected with SARS-CoV-2 have a mortality rate over 20 times higher than their counterparts over 50 years old [
4]. Evidently, the aging process results in a progressive deterioration in pulmonary functionality, increasing vulnerability to lung diseases while compromising the protective barriers of the lung tissue.
The lungs, being the largest organ by surface area in the human body [
5], are critical for gas exchange. Lung function peaks between 18 and 25 years of age and remains stable until around 35 years old. However, subsequent to this period, the progressive aging process gives rise to noticeable alterations in the lungs’ tissue structure, gas exchange function, and immune response mechanisms [
6]. The alveolus, the smallest unit responsible for gas exchange in the lungs, consists of a monolayer of epithelial cells. It serves as a critical site for gas exchange between inhaled air and circulating blood while acting as a protective barrier against harmful substances like pathogens, particulate matter, and other foreign materials [
7]. However, with aging, the protective function of this pulmonary epithelial barrier gradually declines, resulting in reduced responsiveness to environmental stressors. Aging lung tissue exhibits progressive damage and the infiltration of inflammatory cells, accompanied by increased secretion of Senescence-Associated Secretory Phenotype (SASP) and inflammatory factors and the destruction of the alveolar structure. Therefore, effective alveolar regeneration to repair and maintain barrier integrity is crucial for preventing age-related lung diseases.
The epithelial structure is a crucial factor in maintaining alveolar regeneration. The alveolar epithelium principally comprises two distinct cell types: type I alveolar epithelial cells (AT1) and type II alveolar epithelial cells (AT2) [
8]. AT1 cells, characterized by their large and flat squamous morphology, span more than 95% of the alveolar surface, providing an expansive area conducive to efficient gas exchange. Meanwhile, AT1 cells are also the most vulnerable target cells in lung injury and aging. Remarkably, AT1 cells themselves lack the capacity for proliferation. Conversely, AT2 cells serve as stem cells with differentiating capabilities, and their differentiation into AT1 cells is crucial for regenerating epithelial structures. Normally, the bulk of AT2 cells remain quiescent, exhibiting limited regenerative potential [
9]. However, in response to injury signals, AT2 stem cells undergo differentiation and proliferation, ultimately transitioning into AT1 cells to restore the damaged alveolar epithelium [
10]. Previous research revealed that in a bleomycin-induced lung injury model, 12-month-old mice exhibited more severe damage to the alveolar epithelium compared to 3-month-old mice, and during a 63-day observation period, their capacity for alveolar self-repair was weaker than that of the 3-month-old mice [
11]. Aging-related apoptosis, depletion, and the dysfunction of AT2 cells directly impair lung repair and regeneration. The proliferation and differentiation of AT2 cells are critical indicators of alveolar epithelial repair and must be precisely regulated to maintain alveolar integrity and efficient repair [
12,
13]. The disruption of this equilibrium may result in grave pulmonary ailments. The robust vitality and differentiation capacity of AT2 cells are essential for maintaining a sufficient supply of AT1 cells.
As research on AT2 cells progresses, we are uncovering various factors that can impact their self-renewal and differentiation capabilities, including oxidative stress [
14], telomere damage [
15], and mechanical tension [
16], among others. The influence of chronic inflammation resulting from natural aging on the stemness of AT2 cells is increasingly under scrutiny. Chronic inflammation is a persistent state of inflammation that causes tissue damage and may affect stem cell function through multiple pathways. However, it is currently unclear how aging reshapes the microenvironment of lung stem cells and whether the inflammation response induced by aging is a key factor in inhibiting the regeneration of epithelial structures.
In this study, we systematically studied dynamic changes in the quantity, structure, and function of alveolar epithelial cells from lung maturation to senescence, employing a naturally aging mouse model. Additionally, we utilized transcriptomics to further explore the potential factors influencing AT2 function, uncovering a correlation between inflammatory factors and the proliferative and differentiating capabilities of AT2 cells during the aging process. Our study contributes to a comprehensive understanding of the impact of inflammation on alveolar regeneration, offering new insights and directions for delaying or treating age-related chronic pulmonary diseases through systemic inflammation modulation strategies.
3. Discussion
The human alveolar epithelium serves as a critical immune interface and is highly susceptible to damage from external stimuli. AT2 cells play a key role in protecting the lungs against pathogen invasion and host defense, repairing damaged alveoli, and restoring pulmonary homeostasis. Our research findings demonstrate that the alveolar epithelium is highly susceptible to damage as a consequence of aging. During lung aging, there is an age-dependent decline in the quantity of AT2 cells accompanied by structural damage and significant impairments in proliferative and differentiation capabilities. Damaged AT2 cells can activate and recruit immune cells into the alveoli, releasing a large amount of cytokines, thereby rapidly altering the immune microenvironment within the lungs [
28,
29]. It is worth noting that AT2 cells are critical executors responsible for epithelial regeneration. AT1 cells, which are responsible for gas exchange, cannot repair themselves once damaged and rely on differentiation from AT2 cells. The depletion and dysfunction of AT2 stem cells caused by aging can directly impact the regeneration of epithelial structures and the repair of alveoli. Furthermore, our findings demonstrate pronounced inflammatory infiltrates in aged lung tissue and the abnormal activation of inflammatory signals in AT2 cells, consistent with previous research [
5,
30,
31,
32,
33]. The secretion of SASP and pro-inflammatory mediators leads to the formation of an inflammatory microenvironment. Inflammation serves as a beneficial immune response. Nevertheless, as individuals age, their tolerance to antigens and physical, chemical, and other stimuli decreases. Consequently, inflammation may transition to a chronic state of low intensity, resulting in lung tissue dysfunction and cellular degeneration [
34,
35].
The impact of inflammation on the proliferative and differentiation capabilities of AT2 cells exhibits a dual role, with these regulatory behaviors dependent on the type and severity of injury and the inflammatory state. The literature has reported that in the alveolar region, inflammatory responses can regulate the regenerative niche, promoting alveolar regeneration. Organoid screening experiments have revealed that inflammatory cytokines, such as IL-1 and TNF-α, can promote the proliferation of surviving AT2 cells through the IL-1/TNFα-NF-κB signaling axis while preserving their ability to differentiate, therefore promoting lung regeneration after AT2 cell injury induced by influenza [
28]. In the context of chronic lung inflammation induced by cigarette smoke, AT2 cells exhibit a dynamic activation of their stem cell properties, thereby facilitating accelerated proliferation, enhanced differentiation, and augmented resistance to apoptosis [
36]. However, our transcriptomic data revealed a significant negative correlation between inflammatory factors and genes associated with AT2 cell proliferation and differentiation. This suggests that the senescence-mediated inflammatory microenvironment directly inhibits the proliferative and differentiating behavior of AT2 cells. Existing research data also support this finding. During lung regeneration following injury, chronic inflammation mediated by IL-1β hinders the transition of AT2 cells into mature AT1 cells, leading to an abnormal accumulation of the transition-state DATP subpopulation and impaired alveolar regeneration [
19]. During pulmonary inflammation, activated neutrophils have been shown to hinder the proliferation of AT2 cells and induce their apoptosis [
37]. Similarly, studies have found that blunt chest trauma can activate alveolar macrophages and recruit neutrophils to the alveolar region, triggering an inflammatory response and inducing apoptosis in AT2 cells, further exacerbating the detrimental effects of inflammation on lung tissue [
38]. An inflammatory response may activate signaling pathways related to inflammation, including the NF-κB pathway, which alters gene expression patterns and regulatory networks in stem cells [
39]. The upregulation of specific inflammatory genes, like miR-155, might further exacerbate the depletion of stem cell populations [
20]. Inflammatory mediators such as TNF-α and IL-1β have been demonstrated to possess the capacity to impede cell proliferation and induce apoptosis across multiple cell types [
21,
40]. The maintenance of pulmonary immune homeostasis requires the coordinated efforts of lung epithelial cells, local immune cells derived from the bone marrow and lymphatic system, and recruited immune cells [
41,
42]. In aged lung tissues, the disruption of pulmonary immune homeostasis leads to the recruitment of numerous inflammatory factors to the alveolar region. Persistent and prolonged chronic inflammation continually stimulates AT2 cells, culminating in irreversible cellular damage. Additionally, the inhibition of regulatory pathways involved in differentiation impedes the successful transition of AT2 cells into mature AT1 cells, hampering the restoration of a damaged alveolar epithelium and subsequently affecting pulmonary gas exchange. The inflammatory microenvironment induced by aging hinders alveolar regeneration.
In our correlation analysis, we found a significant positive correlation between cell cycle arrest-related genes (such as Tgfb2, Tgfbr1, Tgfbr3, and Tgif1) and inflammatory factors. This indicates that inflammatory factors could hinder the cell cycle of AT2 cells, thus affecting their proliferative capacity. Through further meticulous analyses, we unearthed a compelling negative correlation between key cell cycle regulatory genes, namely Cdk4, Cdk6, and Ccnd3, and inflammatory factors. The proteins encoded by Cdk4 and Cdk6 play a pivotal role in facilitating the transition from the G1 to the S phase of the cell cycle by forming complexes with Ccnd3. Additionally, our research has yielded significant evidence of a positive correlation between the Cdkn3 gene, which encodes the CDK inhibitor, and the majority of inflammatory factors. This inhibition effectively hampers the activity of CDK-related proteins and disrupts cell cycle progression. Furthermore, the protein encoded by Ppard exerts a pivotal influence on cellular proliferation by actively participating in the intricate regulation of cellular metabolism and growth, which displays a significant negative correlation with key inflammatory factors such as Il2, Il4, Cxcl15, and Ccl28. Moreover, we discovered a noteworthy negative association between genes involved in spindle formation and chromosome segregation, including Prc1, Knstrn, Mad2l1, Nusap1, and Bub1, and inflammatory factors. Remarkably, these genes are downregulated in the inflammatory conditions induced by aging, potentially compromising the precise orchestration of mitosis. Intriguingly, we observed significant alterations in transcriptional regulation-related genes during the aging process. In particular, Brca1, which encodes a protein involved in DNA double-strand damage repair throughout the cell cycle; Birc5, which encodes a protein essential for preventing abnormal cell death during cell division; and Lockd, which is responsible for cellular stress response and immune regulation, all exhibited a notable positive correlation with inflammatory factors. Additionally, the proteins encoded by the genes Stat3 and Stat6 are crucial in mediating the signal transduction of various cytokines, thereby regulating cellular immune responses and proliferation. Remarkably, these two genes exhibit positive correlations with most inflammatory factors. The upregulation of these genes further supports the notion that age-related chronic inflammation leads to cellular damage and apoptosis, triggering the activation of defense genes for intracellular regulation. In conclusion, these data indicate that the inflammatory response triggered by aging significantly arrested the cell cycle of AT2 cells, impairing their proliferative capacity and thereby impacting alveolar regeneration.
This study presents a systematic elucidation of age-related structural, quantitative, and functional alterations within the mouse alveolar epithelium, with a particular focus on how inflammatory stimuli guide the fate behavior of AT2 stem cells during lung injury repair subsequent to physiological aging. These findings offer new insights into how chronic inflammation impairs stem cell-mediated alveolar regeneration. Furthermore, we are collectively working toward a more comprehensive exploration of the functional characteristics of AT2 cells in response to inflammation, intending to fully elucidate and delve deeper into the specific regulatory mechanisms underlying this physiological correlation. In the future, there is a promising prospect to utilize systemic inflammation modulation strategies to promote alveolar regeneration and delay or even reverse age-related pulmonary diseases, thereby reducing their incidence and mortality rates among the elderly population.
4. Materials and Methods
4.1. Mouse and Tissue Sampling
All animal experiments were conducted in accordance with guidelines prepared by an animal welfare organization and approved by the Animal Care and Use Committee of China Agricultural University (approval number AW41203202-5-3). Pathogen-free C57BL/6J mice were obtained from Beijing Vital River Laboratory Animal Technology Co., Ltd., Beijing, China and housed individually in a controlled environment (temperature, 22–24 °C; humidity, 60%) with ad libitum access to food and water. Six age groups of mice were included in this study, ranging from 3 to 28 months old (including 3 m, 6 m, 12 m, 18, 24 m, and 28 m), and all mice underwent natural aging without any exogenous stimuli or interventions.
After anesthetizing the mice, lung tissues were harvested by perfusing physiological saline through the heart to remove blood and turn the tissues white. Subsequently, 4% paraformaldehyde (PFA, G1101, Servicebio, Wuhan, China) was perfused for the fixation of the organs and tissues. The lung lobes were dissected, fixed in PFA for 48 h, embedded in paraffin, and sectioned into 5 μm slices. Lung tissues that were not perfused with PFA were rapidly frozen in liquid nitrogen and stored at −80 °C for subsequent protein and RNA analyses.
4.2. HE Staining
The structure of the alveolar epithelium of the lung tissues was assessed using an HE staining kit (G1120, Solarbio, Beijing, China). The lung tissue sections were subjected to staining according to the manufacturer’s instructions. Each age group consisted of six biological replicates. Finally, the sections were examined under a bright-field microscope (DM6B, Leica, Wetzlar, Germany) using a 20× objective lens.
4.3. Immunofluorescent Staining
Tissue sections embedded in paraffin were subjected to deparaffinization and hydration, followed by immersion in sodium citrate antigen retrieval solution (C1031, Solarbio, Beijing, China) and microwave boiling. Afterward, the sections were immersed in a PBS solution containing 1% Triton X-100 (T8200, Solarbio, Beijing, China) for 15 min and blocked with 10% goat serum (C01-03001, Bioss, Beijing, China) at room temperature for 1 h. Primary antibodies were added and incubated overnight at 4 °C. The following day, the sections were incubated with secondary antibodies at room temperature in the dark for 1 h (detailed antibody information is shown in
Supplementary Table S1). The sections were then stained with a DAPI working solution (C0065, Solarbio, Beijing, China) for 10 min and mounted using an anti-fade mounting medium (P0126, Beyotime, Shanghai, Beijing). Observation and photography were performed under a laser confocal microscope (LSM 900/Axio Observer 7, ZEISS, Oberkochen, Germany). Cells grown on glass coverslips were stained using the same method.
4.4. Transmission Electron Microscope (TEM) Detection
Fresh lung tissue samples, measuring approximately 1 mm3 in size, were fixed overnight at 4 °C using a 2.5% glutaraldehyde (P1126, Solarbio, Beijing, China) fixative. Subsequently, the samples were fixed with a 1% osmium tetroxide solution for 2 h. Dehydration was performed using a gradient ethanol series (50%, 70%, 80%, 90%, and 95% for 15 min each), followed by two treatments with pure acetone for 20 min each. The samples were then infiltrated with a mixture of embedding resin and acetone (v/v = 1/1) for 2 h, followed by a mixture of embedding resin and acetone (v/v = 3/1) for 3 h, and finally left overnight in pure embedding resin. The processed samples were embedded, polymerized, and sectioned into ultra-thin slices (70–100 nm). These sections were stained with 2% uranyl acetate and lead citrate for 5–15 min each and observed under a transmission electron microscope (H-7650, Hitachi, Tokyo, Japan). Each group comprised three biological replicates.
4.5. Cell Culture
Human A549 cells were obtained from Procell Life Science & Technology Co., Ltd. (Wuhan, China) and were cultured in Ham’s F-12K medium (PM150910, Procell, Wuhan, China) supplemented with 10% fetal bovine serum (164210, Procell, Wuhan, China) and 1% penicillin–streptomycin (PB180120, Procell, Wuhan, China), following standard laboratory protocols. The average doubling time of the A549 cells was approximately 22 h, indicating their rapid proliferation rate.
Primary AT2 cells were cultured in a specialized medium (CM-M003, Procell, Wuhan, China) fortified with essential growth factors to support their specific requirements. Furthermore, the culture vessels used for the primary AT2 cells were carefully coated with a layer of 5 μg/cm2 of mouse tail collagen (354236, Corning, Bedford, MA, USA) to enhance cell adhesion. All cell cultures were maintained in a dedicated incubator (HERAcellTM 150i, Thermo Scientific, Waltham, MA, USA) set at a controlled temperature of 37 °C with a 5% CO2 atmosphere.
4.6. Cellular Senescence Induction
In order to prevent cellular replicative senescence, we utilized low-passage proliferative A549 cells (<15 passages) in this study. These cells were seeded at a density of 1.5 × 105 cells per well in a 6-well plate and cultured under standard conditions for 24 h to promote firm attachment and proliferation. After this incubation period, each well in the treatment group was supplemented with 2 mL of culture medium containing D-galactose (concentrations: 0, 2, 4, 6, 8, 12, 16, 20, and 30 g/L) for a specified duration (e.g., 24, 48, 72 h), in order to induce cellular senescence. Subsequent staining procedures, as well as extractions of RNA and proteins, were performed on these cells for further comprehensive analysis.
4.7. Cell Viability Testing
In this experiment, we utilized a Cell Counting Kit-8 (CCK-8, CK04, Dojindo, Kyushu Island, Japan) according to the provided instructions. After a certain period of D-galactose-induced senescence in the A549 cells, 10 μL of CCK-8 solution was added, and the cells were incubated at 37 °C in a light-protected, constant-temperature chamber for 60 min. Absorbance at 450 nm was measured using a multi-mode microplate reader (SYNERGYTM HTX, BioTek Instruments, Inc., Winooski, VT, USA). Cell viability (%) was calculated as [(%Treatment − %Blank)/(%Control − %Blank)] × 100%. Each group comprised 6 technical replicates.
4.8. β-Galactosidase Staining
In this study, senescent cells were identified utilizing a commercially available β-galactosidase staining kit (G1580, Solarbio, Beijing, China). The specific experimental operations were carried out according to the method provided in the instructions. The stained senescent cells were meticulously observed and characterized using an inverted microscope (DMi8, Leica, Wetzlar, Germany). Each group comprised three replicates.
4.9. Flow Cytometric Analysis of Cell Cycle Progression
A549 cells in the logarithmic growth phase were inoculated into a 6-well plate at a density of 1.5 × 105 cells per well. Following 24 h of incubation with 30 g/L of D-galactose, the cells were digested to obtain a single-cell suspension. Subsequently, 750 μL of pre-chilled absolute ethanol was gradually introduced, and the cell suspension was placed in a −20 °C freezer overnight for fixation. The next day, the cells were resuspended in 425 µL of Cell Staining Buffer (420201, Biolegend, San Diego, CA, USA), 50 μL of 100 µg/mL RNase (R1030, Solarbio, Beijing, China), as well as 25 µL of Propidium Iodide Solution (421301, Biolegend, San Diego, CA, USA) and incubated on ice in the absence of light for 15 min. Cells were acquired at a low speed, and red fluorescence (PE channel) was detected at 488 nm using a flow cytometer (CytoFLEX, BECKMAN COULTER, Suzhou, China). Each group comprised three replicates.
4.10. Isolation and Culture of AT2 Cells
To prepare a mixture of enzymes for lung tissue digestion, 780 U of Type I collagenase (260 U/mg, 17100017, Gibco, Grand Island, NY, USA), 40 U of Elastase (≥3 U/mg, LS002294, Worthington, Lakewood, CO, USA), and 20 U of Dispase II (0.9 U/mg, 4942078001, Roach, Mannheim, Germany) were dissolved in a total volume of 10 mL of PBS (FB13356, FEIMOBIO, Beijing, China) supplemented with Ca2+ and Mg2+.
The mice were anesthetized, and their lungs were rinsed with a physiological saline solution through cardiac perfusion to remove blood. Following tracheal intubation, the lungs were sequentially injected with 1.0 mL of enzyme solution and 0.6 mL of 1% low-melting-point agarose. The lungs were then removed and placed in the remaining enzyme solution for digestion at 37 °C for approximately 40 min. After digestion, the cell suspension was centrifuged, resuspended in 5 mL of red blood cell lysis buffer (Solarbio, R1010), and incubated on ice for 10 min. Subsequently, the suspension was filtered through a 40 μm cell strainer (SORFA, 251100). The cells were cultured at 37 °C with 5% CO2 for 45 min and then transferred to a new culture dish. This process was repeated three times to eliminate most fibroblasts. Finally, the cell suspension was transferred to a 25T culture flask coated with 5 μg/cm2 of mouse tail collagen (Corning, 354236) and cultured for 24 h before medium replacement.
4.11. Flow Cytometry Analysis and Sorting of AT2 Cells
Lung tissues from 3 m and 24 m mice were enzymatically digested to produce lung cell suspensions, followed by subsequent staining procedures. Within a 500 μL volume of cell suspension, 1 μL of FITC rat anti-mouse CD45 antibody (BD, 553079), PE/Cyanine 7 anti-mouse CD31 antibody (102418, Biolegend, San Diego, CA, USA), APC CD326 (EpCAM) monoclonal antibody (17-5791-80, Invitrogen, Carlsbad, CA, USA), and APC/Cyanine 7 anti-mouse MHCII (I-A/I-E) antibody (107627, Biolegend, San Diego, CA, USA) were added. The cells were then incubated on ice in the absence of light for 30 min for optimal labeling. Subsequently, 5 μL of 7-AAD viability staining solution (420404, Biolegend, San Diego, CA, USA) was added to facilitate the selection of viable cells. Ultimately, the cells underwent a flow cytometry analysis (FACSAriaTM III Cell Sorter, BD, Franklin Lake, NJ, USA) to identify and isolate CD45− CD31− CD326+ MHCII+ cells, specifically sorted as AT2 cells.
4.12. RNA Extraction and RT-qPCR
The RNA extraction process involved the lysis of the lung tissue samples and cells using pre-chilled TRNzol reagent (DP424, TIANGEN, Beijing, China) for 30 min, followed by RNA isolation as per the manufacturer’s protocol. RNA concentration was determined using a NanoDrop spectrophotometer (Thermo Scientific, Madison, WI, USA). Subsequently, cDNA synthesis was performed using the cDNA Reverse Transcription Kit (G592, abm, Zhenjiang, China). For a gene expression analysis, an RT-qPCR was conducted on a real-time PCR instrument (QuantStudioTM 5, Thermo Scientific, Marsiling Industrial Estate, Singapore), using SYBR Green (A25742, Thermo Scientific, Madison, WI, USA) as the fluorescent dye. Each group comprised 6 biological replicates, with 3 technical replicates. The data were presented as fold change values calculated using the ∆∆Ct method, with GAPDH used as the reference gene for normalization. The primer sequences used in this study are provided in
Supplementary Tables S2 and S3.
4.13. Western Blots
For lung tissue samples, approximately 80–100 mg of fresh tissue was incubated on ice for 30 min with 1 mL of lysis buffer containing 980 μL of RIPA (R0010, Solarbio, Beijing, China), 10 μL of PMSF (P0100, Solarbio, Beijing, China), and 10 μL of protein phosphatase inhibitor (P1260, Solarbio, Beijing, China) and underwent homogenization and centrifugation. For monolayer adherent cells grown in a 6-well plate, 1 mL of lysis buffer was added to each well to obtain the protein supernatant. SDS-PAGE was utilized to separate the high-molecular-weight proteins in each sample. Incubation with specific antibodies against the target proteins was performed (Detailed antibody information is shown in
Supplementary Table S4), and the bound antibodies were detected and visualized using a chemiluminescence imaging system (ChemiScope 6100, Clinx Science Instruments, Shanghai, China). Each group comprised three replicates.
4.14. RNA Sequencing Analysis
AT2 cells were isolated from the lungs of both 3 m and 24 m mice using flow cytometry (FACSAriaTM III Cell Sorter, BD, Franklin Lake, NJ, USA). Three biological replicates were collected for each age group by pooling lung cells from multiple mice to ensure an adequate number of AT2 cells for subsequent sequencing. A transcriptome sequencing analysis was conducted on the Illumina NovaSeq 6000 platform (sequencing mode: PE150) at Shanghai Applied Protein Technology, Shanghai, China.
4.15. Statistical Analyses
The data were presented as mean ± SD values or in violin plot form, with n representing the number of experimental replicates or the number of mice. The data analysis was conducted using IBM SPSS Statistics version 26.0 software. When comparing differences between two groups, t-tests were employed to calculate p-values. Statistical significance was indicated by asterisks, where * p < 0.05 and ** p < 0.01, while “ns” denoted non-significance. For comparisons among multiple groups, post hoc multiple comparisons using Duncan’s analysis within a one-way ANOVA test were performed, and lowercase letters were assigned to indicate statistical differences. In all cases, p < 0.05 or p < 0.01 was considered statistically significant. Graphical representations of the analyzed data were generated using GraphPad Prism version 9.0 software.