1. Introduction
The occurrence of the novel coronavirus disease 2019 (COVID-19) pandemic caused by SARS-CoV-2 mostly affected the aged and immunocompromised population. Millions of lives were lost because of the virus and posed a new challenge for the medical sciences. The epidemiology, molecular properties, processes, clinical manifestations, and consequences of this viral infection are the subject of thousands of investigations. Still, there is a possibility of new emerging strains that may be deadlier than the ones we already have. Primarily, SARS-CoV-2 causes hyperinflammation and acute respiratory distress syndrome (ARDS) in the lungs via the respiratory tract. ARDS may further result in organ damage because of micro-/macro-thromboembolism, aberrant complement activation, or extended viremia, which may lead to systemic multiple organ dysfunctions [
1]. Patients who already suffered from COVID-19 may experience post-acute consequences, and certain symptoms may last for several months. According to the World Health Organization (WHO), long-COVID symptoms could arise from COVID-19-induced hypoxia, decreased cardiac output, myalgia, breathlessness, or chronic fatigue syndrome-like symptoms. The possibility of developing neurodegenerative disorders, cancer, and cardiovascular disorders are a few examples of the long-term negative effects that may occur [
2]. According to the literature, these symptoms are a direct result of redox imbalance, excessive free radical formation, mitochondrial degeneration, and decreased oxidative phosphorylation [
3].
The literature suggests that SARS-CoV-2 causes COVID-19 pathogenesis through its protruding spike (S) glycoprotein which interacts with the host cell receptor angiotensin-converting enzyme 2 (ACE2). When the ACE2 receptor interacts with the virus via non-covalent interactions, the downstream events take place [
4]. Severe symptoms of COVID-19 accompany a process often referred to as a “cytokine storm”, where extensive inflammation takes place because of the uncontrolled release of pro-inflammatory cytokines, growth factors, and interferons; encompasses an expanding tropism of SARS-CoV-2 for vital organs like the heart, kidney, pancreas, etc.; and induces many systemic abnormalities. As a result, a chronic pathological condition becomes established, known as long COVID. During a cytokine storm, prominent rises in serum inflammatory cytokine levels, such as interferon-γ, IL-6, IL-10, IL-8, tumor necrosis factor (TNF) α, blood ferritin level, and soluble IL-2 receptor alpha, are usually present [
5].
There are studies where individual structural proteins of SARS-CoV-2 were administered to understand their antigenicity. The S protein induces a variety of inflammatory changes in innate immune cells, e.g., macrophages, monocytes, and neutrophils [
6]. Mechanistically, the S protein of SARS-CoV-2 plays a key role in the process of receptor recognition and cell membrane fusion process. It is made up of two subunits, S1 and S2. The S1 subunit contains a receptor-binding domain that recognizes and binds to the host receptor ACE2, which is usually present on epithelial cells of the alveoli, thereby causing viral endocytosis [
7]. The immune cells are triggered by viral constituents and by-products of apoptotic as well as necrotic cells, causing the release of inflammatory mediators, and thus hyperinflammation and thrombus in SARS-CoV-2-infected lungs.
The pathogenesis of COVID-19 infections is significantly aided by the development of oxidative stress in alveolar epithelium and endothelium. Though a basal level of reactive oxygen species (ROS) production is required for targeting infection, the excessive production of ROS and low concentration/activity of intrinsic antioxidants creates an imbalance, and consequently oxidative stress, which results in cell and tissue damage [
8]. Concurrent inflammation and thrombosis promote the excessive formation of ROS, resulting in a self-sustaining cycle between oxidative stress, cytokine storm, and disease progression [
9]. The overproduction of mitochondrial ROS during inflammatory reactions in alveolar cells results in further downstream signaling. Pro-inflammatory cytokines and oxidative stress markers are linked in patients with severe COVID-19, which is supported by the literature [
10,
11]. In addition, disruptions in ATP synthesis, the electron transport chain, and any malfunctions in the opening of the mitochondrial permeability transition (MPT) pore can stimulate the apoptotic pathway of the mitochondria. Mechanisms like alterations in transmembrane potential and oxidative damage to the mitochondrial DNA, proteins, and membrane phospholipids can also contribute to the above pathway. When mitochondria are damaged, cytopathic hypoxia develops, which causes partial oxygen reduction and ROS formation, resulting in damage to the outer membrane and release of cytochrome C, and eventually cell death and multi-organ failure [
12].
Finding a precise treatment for COVID-19 is crucial as the pandemic relapses repeatedly. Moreover, the long-term negative consequences of the existing vaccinations that have been developed to target different SARS-CoV-2 proteins are still unknown [
13]. In this context, improvements can be greatly sourced from natural sources. The antiviral actions of various polyphenols from herbal preparations against viral diseases were explored, and interesting outcomes were published; however, the precise mechanisms and pathways of these formulations are still obscure. The ultradiluted formulation of
Eupatorium perfoliatum (UDE), an extract from a naturally occurring plant that has historically been used as alternative therapeutics for fever and infections, was used in this study. Past investigations demonstrated that the
Eupatorium perfoliatum extract has antiviral properties against influenza A virus [
14]. Previously, researchers from our laboratory demonstrated its role against Dengue virus infection with a precise mechanism of action [
15,
16]. Herein, we diluted the tincture of
Eupatorium perfoliatum according to the Pharmacopoeia of India, 2016, and further diluted using one part of the tincture and nine parts of 70% ethanol. The preparation of subsequent dilutions was carried out with 10 μL of the tincture solution and 990 μL of 70% ethanol up to six times. Then, we investigated its action responses in reducing inflammation caused by recombinant S protein both in cultured cells and BALB/c mice. Our previous study on S protein induction in BALB/c mice showed the remodeling of the extracellular matrix and the role of matrix metalloproteinase-9 (MMP-9) during lung fibrosis [
17]. We put forth a hypothesis supported by substantial scientific evidence that links oxidative stress to mitochondrial malfunction and explained how it contributes to the exacerbation of the cytokine storm. This study documented the potential role of UDE in mitigating the inflammatory response and ROS-mediated oxidative stress to resist the cytokine storm and subsequent apoptosis during S protein-induced pathogenesis. UDE exhibited significant potential to alleviate mitochondrial dysfunction with reduced toxicity and concurrent apoptosis in the lung caused by S protein. The study from this research can also direct future research and treatment plans with UDE in other respiratory diseases.
2. Materials and Methods
2.1. Chemicals
SARS-CoV-2 recombinant S protein (Catalogue no. RPO1260LQ) and ELISA kits were procured from Abclonal, MA, USA. Cell culture media, phosphate-buffered saline (PBS), and fetal bovine serum (FBS) were purchased from Invitrogen, Thermo Fisher Scientific (MA, USA). Acetaldehyde-free ethanol and the tincture solution of UDE (Batch no. 0711) was obtained from HAPCO, West Bengal, India. The MTT reagent (Catalogue no. MTT-TC191) and 4,6-diamidino-2-phenylindole (DAPI) reagent (Catalogue no. TC229-5MG), cell culture plasticware, and dimethyl sulfoxide (DMSO) were purchased from HiMedia (Mumbai, India). Fine chemicals, includingprimary and secondary antibodies, the substrate, and the cellular ROS generation kit, were acquired from Santa Cruz Biotechnologies Inc. (TX, USA), Life Technologies (MA, USA), Abclonal (MA, USA), and Sigma-Aldrich Co. (St. Louis, MO, USA).
2.2. Reconstitution of SARS-CoV-2 Recombinant and Spike Antigen
SARS-CoV-2 S1 + S2 ECD (S-ECD) recombinant protein labeled with His tag (wild type, produced by cloning in HEK293 cells, which were endotoxin-free and appeared as a band in SDS-PAGE at 35 kDa) was reconstituted in 1 mL sterile distilled water as per the manufacturer’s data sheet and filtered. It was further dissolved in sterile PBS (pH 7.4) for dilution, and finally, a solution was prepared with a concentration of 2.5 µg/mL, which was used in the experiments to mimic the COVID-19 pathogenesis. Endotoxin tests using an E-toxate kit (Sigma-Aldrich, MO, USA), microbial tests, and microscopic examinations were performed to check the presence of any contamination in the prepared tincture.
2.3. Cell Culture
The murine macrophage cell line, RAW 264.7, was purchased from National Centre for Cell Science, Pune, India. Cells were preserved in the laboratory in an incubator with a 5% flow of CO2 at 37 °C and 95% air. These were grown and maintained in MEM media, containing 10% (by volume) FBS and 1% antibiotic solution. For experiments, cells were seeded (2 × 106 cells/well) into 6-well plates and cultured for 24 h. Serum-free conditions were used to avoid potential drug–protein interactions.
The following groups were included in all cell culture experiments and conducted in triplicates: (i) untreated control, (ii) only S protein treated, (iii) UDE + S protein treated, and (iv) S protein + UDE treated.
2.4. In Vitro Cytotoxicity by MTT Assay
The cytotoxicity of S protein and UDE was determined via colorimetric MTT assay. RAW 264.7 cells (1 × 104 cells/well) were cultured in a 96-well plate under optimum conditions. Once the cells were confluent, different wells were treated with varied concentrations of S antigen solution, having a stock concentration of 2.5 µg/mL (0.5–3% of the media volume) for varying periods. In the same way, UDE, having a stock concentration of 100 pg/mL, was applied to the cells (0.5–4% of the media volume) for the standardization of concentration and time of treatment. The experiment was conducted in triplicates, and blank wells contained only the medium and no cells. A graph consisting of dose versus time was prepared to standardize the doses of S protein and UDE.
2.5. Wound Healing/Cell Scratch Assay
The impacts of S protein and UDE on the migration or invasiveness of RAW 264.7 cells were examined using this approach [
18]. Cells were seeded in 6-well plates and maintained until they attained 85–90% confluency. The growth media was then changed without FBS for 6hbefore administration of either UDE or S protein. Using a sterile p-200 pipette tip, a gap, or “scratch”, was formed on the confluent cellular monolayer. At this point, images of the fields were captured and are referred to as 0 h images. Cells were gently rinsed with PBS. After that, pre- and post-treatment with UDE and S protein were conducted in IC
50 concentrations, respectively, obtained from the MTT assay, and a serum-free condition was maintained for a further 24 h of incubation. An Olympus inverted microscope with a digital camera was used to study the gap’s closing, and pictures were obtained at 10× magnifications. The size of the wound and its closure were determined using Image J software (V 1.54, NIH, Bethesda, MD, USA), and proliferation potential was calculated as follows:
2.6. DAPI Staining to Analyze Nuclear Morphology
RAW 264.7 cells were inoculated into 6-well plates and incubated for 24 h. Then, the cells were treated with S antigen at its IC50 concentration, and the preventive and curative administration of UDE was carried out via incubation for another 6 h. Following the fixation of cells with methanol, they were stained with DAPI (10 µmol/L working solution) and incubated for 30 min in the dark. After washing twice in PBS cells, they were observed under an Andor confocal microscope.
2.7. Immunofluorescence
RAW 264.7 cells were grown on glass coverslips. After treatment, they were fixed in 2% paraformaldehyde in PBS. Then, 0.2% (by volume) Triton X-100/PBS and0.1% (w/v) bovine serum albumin (BSA) were used to treat the cells. Next, the cells were treated with primary antibody for Caspase-3 (1:200 dilution), washed, and then incubated with the secondary antibody that was conjugated with rabbit anti-goat FITC. Coverslips were mounted and magnified 60 times using an Andor confocal microscope. The fluorescence intensity of individual pictures was analyzed using Image J.
2.8. Animal Experiment
The study procedures were approved by the Institutional Animal Ethics Committee, (Sanction no. DACRRIH/CPCSEA/IAEC/2021/08). For the experiments, adult BALB/c mice (male and female)with an approximate individual weight of 25–30 g were taken. These mice were kept in the animal house for a week to acclimatize to the conditions of 60 ± 10% relative humidity, 21 ± 2 °C, and 12 h/12 h photoperiod). Before the experiments, animals were randomized by a veterinarian who was independent of study conduct. An acute toxicity study was performed with female mice to know the median lethal dose (LD50) of UDE according to the OECD 423 guideline. The dosing of animals and the observation of clinical signs were recorded in a blind manner. There was zero mortality at all tested levels of doses and LD50 of UDE upon a single oral administration, per Globally Harmonized Classification System, and the doses belonged to category 5 or were unclassified (LD50 cut off is ∞).
The mice were randomly allocated into 4 groups (6 mice/group) as follows: (i) control (oral administration of water only for 3 consecutive days), (ii) treatment with only S-protein intranasally (5 µL from the stock concentration of2.5 µg/mL) for 3 consecutive days, (iii) treatment with UDE orally (30 µL to each animal)once daily for 3 consecutive days followed by S-protein intranasally for 3 consecutive days, and (iv) treatment with S-protein intranasally for 3 consecutive days followed by UDE orally (30 µL to each animal), once daily for 3 consecutive days. Ad libitum food and drinking water were provided to them throughout the treatment period except for acute oral toxicity test. A cocktail of ketamine (80–100 mg/kg IP) and xylazine (10–12.5 mg/kg IP) was used for euthanasia for each animal. Lung tissues of each mouse were uniformly inflated using intratracheal instillation of 10% formalin using a 22 g needleat a rate of 200 µL/second until there was a reflux. After 7 days of the last dose administration, all animals were sacrificed; blood by puncturing retro-orbital sinus and lungs were collected. Sera from blood were separated and used to measure the biochemical profile of lungs, as well as antioxidant and oxidative stress markers.
2.9. Histology, Immunohistochemistry (IHC), and Immunofluorescence (IF)
For histological examination, a microtome was used to prepare tissue sections of 4–5 µm thickness from paraffin-embedded blocks, which were stained with hematoxylin and eosin. The prepared slides were observed using a light microscope (Olympus, Tokyo, Japan) for histopathological alterations.
The same unstained tissue sections were utilized to verify the localization of MMP-9 and iNOS by IHC. Sections were washed, 1% trypsin in PBS was applied for unmasking antigens, and the endogenous peroxidase activity was blocked by 3% H
2O
2. After blocking non-specific sites with 5% BSA in Tris-buffered saline (TBS) (20 mM Tris-HCl, pH 7.4 containing 150 mM NaCl), tissue sections were incubated with primary antibodies (Santa Cruz Biotechnology, Inc., Dallas, TX, USA) diluted in TBS, overnight at 4 °C, and washed thoroughly. Secondary biotinylated antibodies were used to detect the binding of the primary antibodies: goat anti-mouse (Santa Cruz Biotechnology, USA). Next, horseradish peroxidase (HRP)-conjugated avidin and 3,3-diaminobenzidine (DAB)–H
2O
2 were used to develop the reaction. Tissue sections were counterstained with Mayer’s hematoxylin and examined using an Olympus microscope. Images were captured with CellSeNS Entry software, V: 1.15 [
19,
20].
The localization of cytochrome C was checked by IF using the same unstained lung tissue sections. Antigen retrieval from the tissue sections was performed using trypsin (0.05% trypsin, 0.1% CaCl
2). Followed by blocking with 5% BSA and extensive washing with TBS, the sections were incubated with primary antibodies (Santa Cruz Biotechnology, Inc., Dallas, TX, USA), diluted in TBS, kept overnight at 4 °C, and washed thoroughly. A solution of the FITC-conjugated secondary antibody (Santa Cruz Biotechnology, TX, USA) was then incubated with the sections, and further counterstaining with DAPI was performed. The resultant sections were washed meticulously with PBS, and images were documented using confocal microscope, Andor Technology (Belfast, Northern Ireland, UK) [
12].
2.10. ELISA of Cytokines
The cytokines and relevant signaling molecules, caspase-1, iNOS, IL-6, and IL-18 were measured using ELISA kits from ABclonal. Standard curves for the molecules were prepared as directed in the literature [
21]. Absorbance was measured using an ELISA reader (Bio-Rad Laboratories, Hercules, CA, USA) at 570 nm as per the manufacturer’s instructions.
2.11. Mitochondrial ROS Measurement
Mitochondria from mice lung tissues of different groups of animals were isolated using the differential centrifugation method. Fluorescence intensity was measured using a spectrofluorometer (LS3B, Perkin Elmer, MA, USA) with wavelengths of 499 nm and 520 nm as excitation and emission wavelengths, respectively [
12]. The data were normalized to normal values, with 100% representing anormal value.
The intracellular ROS level in RAW 264.7 cells was calculated with Life Technologies kit, and micrographs were captured using a confocal microscope. The experiment was based on the uptake of fluorogenic marker 5-(and-6)-carboxy-2′,7′-dichlorodihydrofluoresceindiacetate (carboxy-H2DCFDA) in live cells. The fluorescence intensity of each image was evaluated using Image J software.
2.12. Succinate Dehydrogenase (SDH) Activity Assay
Based on the reduction of DCIP by phenazine methosulphate (PMS), SDH activity was analyzed spectrophotometrically. Notably, 1 mL of final reaction mixture contained 0.01 mL of diluted enzyme + 0.78 mL of 50 mM Tris-HCl (pH 8.2) + 0.1 mL of 1.5 mM DCIP (10 mM KCN + 0.1 mL of PMS of 1 mg/mL). The absorbance of DCIP was measured at 600 nm after the reaction was initiated with 0.01 mL of 0.5 M sodium succinate (pH 8.0). One unit of enzyme activity corresponded to the quantity of enzyme that reduced 1 μmol of DCIP per min, withan extinction coefficient of 21 mM
−1cm
−1 [
22].
2.13. Activity Assay for NADH Oxidase
NADH oxidase activity was assayed spectrophotometrically in a solution of pH 7.4 consisting of 50 mM Na
2HPO
4–KH
2PO
4. The reaction began when 0.125 mM NADH was added to the preparation of sub-mitochondrial particles (0.2 mg/mL). Enzyme activity corresponded to the oxidation of NADH, which was measured at 340 nm (ε = 6.22 mM
−1cm
−1) [
23].
2.14. Assay for Lipid Peroxidation
Lipid peroxidation corresponded to the quantity of conjugated diene in the lung epithelial cells which was measured using a spectrophotometer. Lung tissue homogenates were prepared to extract mitochondrial membrane lipid using a chloroform–methanol mixture (2:1,
v/
v). The extract was allowed to evaporate to a dry powder in a nitrogen atmosphere at 25 °C, followed by dissolving in n-cyclohexane. Cyclohexane containing lipids were measured at 234 nm, and the results were quantified as micromoles of lipohydroperoxide/mg of protein where ε is 2.52 × 10
4/M/cm [
24]. Total protein estimation was carried out by the Lowry method [
25].
2.15. Myeloperoxidase Assay
Myeloperoxidase (MPO) enzyme activity was quantified spectrophotometrically using guaiacol as a substrate. The reaction mixture was made with 0.5 mM H
2O
2 + 0.4 M guaiacol in phosphate buffer (50 mM, pH 7.4). Then, 1 mL of the reaction mixture was taken, and lung tissue homogenate was added to observe any alterations in the per-minute absorbance of tetraguaiacol at 470 nm using a Multiskan Sky (Thermo Fisher Inc., MA, USA) [
26].
2.16. RNA Extraction and Semi-qRT-PCR
The lung tissue samples from mice were transferred to vials containing 1 mL of TRIZOL (Ambion, Life Technologies, MA, USA), which were homogenized. The total RNA from lung tissues was extracted according to the kit instructions. By measuring the total RNA at 260/280 and 260/230 nm using a spectrophotometer, the amount of total RNA was quantified.
For each sample of total RNA, reverse transcription was performed as per manuals of the cDNA reverse transcription kits iScript (Bio-Rad Laboratories, CA, USA). The PCR products were separated using 1% agarose gel stained in ethidium bromide and observed under a transilluminator. Relative expressions of mRNAs between different groups of animals, as mentioned in
Section 2.8, were compared in quantitative real-time PCR (Bio-Rad CFX96, Singapore). The alterations in mRNA expression were normalized as a fold change with respect to the normal control, and the same was compared to
glyceraldehyde 3-phosphate dehydrogenase (
GAPDH). The NCBI blast and SnapGene PCR primer designing platforms were used to create all of the primers for the DNA oligonucleotides [
16].
2.17. Western Blot
Total proteins were extracted from the tissues with a lysis buffer, followed by the measurement of protein concentrations by the Lowry method [
24]. After quantitation under different concentrations and protein separation by polyacrylamide gel electrophoresis, proteins were transferred to polyvinyldiene fluoride membranes through the semi-dry transfer method and blocked for 1 h in 5% BSA. Next, the primary antibodies were dropwise added on the membranes, superoxide dismutase1 (SOD1), iNOS, bcl-2, bax, and caspase-9 incubated overnight at 4 °C. After 3 washes with TBST, HRP-labeled secondary antibodies were added. After 3 TBST rinses, the chemiluminescence solution was added for development using ChemiDoc (Bio-Rad, Laboratories, CA, USA). Individual bands were analyzed for their pixel density using Image J software [
12].
2.18. Data Interpretation and Statistical Analysis
The data in the graphs are represented as the mean ± standard error. To determine the mean and standard deviation, all data were collected in duplicate and three copies. An analysis of variance was used to see whether there were significant variations between the means. MTT assay, ELISA, and RT-PCR data were analyzed using Sigmaplot. As stated in the text, the Student–Newman–Keuls test (ANOVA) and Student’s t-test were used for the statistical analysis. All data analyses were performed using the statistical software Sigmaplot version 10.0. When p value < 0.05, a difference was deemed significant.
4. Discussion
The attempt to reduce the impact of the COVID-19 disease burden is an important concern for the research community and global healthcare. To gain insights into the mechanisms of disease progression, virus evasion strategies, and host responses and to create efficient antiviral therapeutics, animal models mimicking viral infection and subsequent pathogenesis are extremely important. Several critical lung disorders, including COVID-19 and ARDS, may lead to pulmonary fibrosis, destroy alveolar shapes, and impair lung function. But, before proceeding to animal models, cell-based models were utilized, and the results were further extrapolated to BALB/c mice for validation [
29]. To mimic the general physiological and pathological processes, we used mouse macrophage cell line RAW 264.7 and administered S protein in vitro to assess the preventive and therapeutic effects of UDE. This cell line is a well-known in vitro disease model for investigating the bioactivity of therapeutic agents and their efficacy, which are indicative of the potential response in human cells. Wang et al., 2022, described the role of alveolar macrophages as first-line defenders that scavenge the inhaled pathogen or xenobiotics to maintain lung homeostasis [
30]. Studies demonstrated that viruses not only infect alveolar macrophages but also replicate in the lower respiratory tract, causing cytotoxicity, overproduction of pro-inflammatory cytokines and interferons, and finally, fatal pneumonia [
31]. From the histological pictures, we showed the infiltration of inflammatory cells and macrophages in the lung of S protein-treated group of mice.
ROS and reactive nitrogen species (RNS) in cells and tissues are a double-edged sword because they can affect physiological function both favorably and unfavorably. Beyond the threshold level, ROS/RNS damage membrane lipids, proteins, or DNA, thus leading to biological damage. Out of several ROS-generating systems, macrophage stimulation in the alveolus and mitochondrial degeneration induced the overproduction of ROS in mitochondria via either the electron transport chain or cytochrome P450 as a metabolic by-product. The mitochondrial respiratory chain is primarily responsible for the overexpression of ROS, which, in turn, produces potential oxidants such as superoxide, H
2O
2, and other reactive species. H
2O
2 causes the expression of pro-inflammatory cytokines, such as IL-6, IL-1β, and TNFα, as well as iNOS, through a redox-sensitive transcription factor, NF-κB [
32]. In the present study, qRT-PCR data showed that the transcriptional expressions of IL-1β and IL-18 were significantly high in response to the S protein. A similar trend was reflected in ELISA where the circulating level of IL-18 was found to be very high in the S protein-treated group. However, IL-1β in mice serum was below the detectable range during ELISA, probably because of its extremely short half-life in the circulatory fluid [
33]. So, it may be hypothesized that NF-κB acted as a transcription factor for synthesizing IL-1β and IL-18. In response to the S protein, mRNA levels of inflammatory cytokines like IL-6, TNF-α, etc., in the lungs of BALB/c mice substantially increased. IL-10 played a paradoxical role in this study. The mRNA expression of IL-10 was upregulated with S protein administration. It was found further upregulated upon the therapeutic administration of UDE in both male and female mice, which probably suggests that it played an anti-inflammatory role in curbing the inflammatory response, unlike IL-6 and other pro-inflammatory cytokines.
According to studies, the S protein of SARS-CoV-2 interacts with the host either through ACE2 receptors with the help of TMPRSS2 or by other mediators like TLR-4, or TLR-2, and cellular stress is induced [
34]. It further activates macrophages and neutrophils, elevates IL-6, IL-10, and other inflammatory markers, and contributes to the development of acute pulmonary inflammation [
35].
UDE was able to bring down the transcriptional levels of both IL-1β and IL-18 and the translational level of IL-18. MMP-9 is a downstream target of the NF-κB signaling pathway, which was also upregulated, implying ECM remodeling during S protein-mediated lung injury. So, increased NF-κB levels were closely linked to the expression of MMP-9 and the emergence of the cytokine storm during the progression of the disease [
36]. Mounting evidence also points to a direct connection between the production of ROS and the expression of iNOS, and consequently, the activation of the cytosolic multiprotein complexes and sensors of the innate immune system called “inflammasome” and further downstream signaling [
37]. In this study, RT-PCR, ELISA, and Western blot data confirmed the significant upregulation of iNOS and caspase-1 in both the transcriptional and translational levels in response to S protein. Caspase-1 serves an essential function in the initiation of inflammation by the efficient cleavage and maturation of cytokines, IL-1β and IL-18, from their inactive precursors, both of which play important roles in host defense and mediate the pathogenesis during COVID-19 infection. RT-PCR also confirmed the significant transcriptional upregulation of these two cytokines which further downregulated with the therapeutic administration of UDE.
The activation of inflammasome resulted in excessive levels of cytokine production, which is referred to as cytokine storm and triggers a multifaceted and aberrant uncontrolled response. It involved pro-inflammatory cytokines, notably IL-6, IL-1β, and TNF-α. These cytokines, in turn, activated more macrophages, neutrophils, endothelium cells, and other immune cells to produce more superoxide, peroxynitrite, and free radicals which are harmful to mitochondria. We performed RT-PCR on all these cytokines, which appeared excessively upregulated in the S protein-treated group of mice. Interestingly, they all were downregulated in both male and female mice when UDE was administered orally.
Although the inflammasome and oxidative stress were both independently associated with COVID-19, it is unknown if these two mechanisms combine to increase disease severity. Here, we discovered a link between the elevated levels of SOD1 (through Western blot) and lipid peroxidation (spectrophotometrically), two characteristics of the oxidative stress response, in lungs treated with S protein, which substantially linked with high caspase-1 activity and excessive ROS formation. The literature also showed that the antioxidant enzyme SOD1 regulates both ROS in the cytoplasm and caspase-1 activation [
38]. In this study, Western blot, as shown in
Figure 8, depicted significant upregulation in SOD1 in S protein-treated tissues but again downregulated when UDE was applied to mice. Apart from the cytokine maturation or formation of inflammasome complex, caspase-1 regulates multiple cellular functions like inflammatory response, regulation of protein cleavage, apoptosis, etc. [
39].
Increased ROS formation also leads to the rupture of mitochondria and the release of mitochondrial DNA. The release of mitochondrial DNA facilitates inflammatory response via the activation of the inflammasome. The consequences of mitochondrial damage are decreased ATP supply and cell death. The overall mitochondrial damage includes loss of size, shape, and integrity. It also may result in lowered membrane potential formation of MPT pore formation, leading to swelling. Additionally, S protein inhibited antioxidant enzymes’ ability to defend the mitochondria. NADH oxidase and succinate dehydrogenase activities are present in the mitochondrial respiratory complex I and complex II, respectively. A higher generation of ROS is suggested by increased NADH oxidase activity. Cytochrome C release is linked to the impairedelectron transport mechanism in mitochondria which facilitates apoptosis. Reduced SDH activity in lung epithelial cells also indicates less proton accumulation, which dissipates the potential of the mitochondrial membrane and allows the release of cytochrome C into the cytosol, which is confirmed in
Figure 10. Our study clearly showed that UDE potentially resisted the excess ROS formation, both in vitro and in vivo; thereby inhibiting mitochondrial degeneration, leaking cytochrome C, and further downstream apoptotic events through caspases.
In our study, we noticed a significant upregulation of IL-6, IL-10, IL-1β, TNFα, iNOS, MMP-9, NF-κB, and MPO in response to S protein, which are the contributors to acute inflammatory response. The histopathology analysis of S protein-treated lungs of mice showed a substantial accumulation of inflammatory cells, lesions such as edema, inflammation, and a change in alveolar septal thickness. An elevated level of free radicals accompanies the over production of cytochrome C, which exacerbates the tissue damage and alters the redox equilibrium of the lungs.
The intranasal administration of UDE substantially reduced the permeation of inflammatory cells in the lungs of BALB/c mice. UDE demonstrated its antioxidant capability by reducing lipid peroxidation, protein carbonylation, and MPO activity during the healing of lung inflammation. Additionally, the cytoprotective effects of UDE were linked to the downregulation of inflammatory cytokines and restoring the homeostasis of free radicals and antioxidants. UDE administration provided significant protection in retaining the shape and integrity of mitochondria by regulating the cytoplasmic release of cytochrome C, and excess ROS production. The prophylactically UDE-treated group showed almost no localization of cytochrome C compared with the control, which indicates mitochondrial regeneration. On the other hand, the overproduction of ROS can also activate pro-apoptotic Bcl-2 family proteins by releasing cytochrome C from damaged mitochondria. It is detected by APAF-1 and pro-apoptotic caspase-3 and -9 [
40,
41]. The degree to which the apoptotic pathway is activated during COVID-19 has a direct association with the development of multiple complications. According to published research, the complex apoptosis process is influenced by caspases and the Bcl-2 protein family [
42,
43]. In the cascade of protease cleavage during apoptosis, the primary function of caspase-3 is to eliminate damaged cells. After activation, it cleaves proteins required for cell viability, thereby triggering apoptosis. When we tried to see the tissue damage in light of apoptosis, we found significant upregulations of caspase-3 and Bax, at both the transcriptional and translational levels, and the downregulation of Bcl-2 in the S protein-treated mice. RT-PCR data confirmed that therapeutic use of UDE reduced inflammation by normalizing the expression of those pro-apoptotic markers, increasing the expression of anti-apoptotic proteins like Bcl-2, scavenged ROS, and protecting the lung from oxidative damage. The terminal event of S protein-induced hyperinflammation is apoptosis, which results in cell lysis, the release of cytosolic contents to the extracellular space, along with tissue damage. In summary, the therapeutic administration of UDE was more effective against S protein-mediated damage. It probably inhibited hyperinflammation, mitochondriopathy, and apoptosis by controlling the expression of individual mediators of the signaling pathway, there by reducing the lung damage caused by the S protein. Nonetheless, UDE prevented the S protein mediated in many instances.