COVID-19 is a serious infectious disease that spreads globally and threatens the health and lives of people worldwide since the end of 2019. It was reported that low plasma 25(OH)-vitamin D (VitD) level is an independent risk factor for COVID-19 incidence and hospitalization33. On the other hand, supplement of VitD in the treatment regimen for COVID-19 patients achieved certain therapeutic effects and reduced inflammatory markers34. And the immunomodulatory function of VitD was effective in preventing multiple organ failure, cardiovascular complications and other side effects caused by cytokine storm in COVID-19 patients35. These evidence implicate a close association of VitD insufficiency to the pandemic and clinical manifestations of the COVID-19. It is well known that VitD insufficiency is also the key causative factor for osteoporosis and VitD is one of the major therapeutic agents in the treatment of osteoporosis[10]. Indeed, osteoporosis patients were found to be more susceptible to SARS-COV-2 infection and osteoporosis manifestations became worse after suffering from COVID-19 while some COVID-19 patients developed osteoporosis as a complication36. Therefore, VitD insufficiency may be a risk factor for both osteoporosis and COVID-19. VitD has been suggested as a potential adjuvant or alternative medicine for both osteoporosis and COVID-1912. It is currently unknown, however, whether and how VitD insufficiency links osteoporosis with COVID-19. In this study, we used multiple approaches, including network pharmacology, bioinformatics, and molecular docking to identify and characterize the VitD targets on COVID-19 and osteoporosis and to explore the molecular mechanisms for the potential therapeutic effects of VitD in the treatment of both COVID-19 and osteoporosis. The major novel findings in this study include: 1)among a total of 243 VitD targets 42 common targets on both COVID-19 and osteoporosis were identified; 2) in the VitD-COVID-19-osteoporosis network EGFR, AR, ESR1, MAPK8, MDM2, EZH2, ERBB2 and MAPT were determined as core targets; 3) VitD binds tightly and interacts physically with EGFR and MAPK8; 4) these VitD targets involved in the ErbB and MAPK signaling pathways are critical for the regulation of lung fibrosis, bone structural integrity, and immune responses. These findings provided novel mechanistic insights into the functional roles and molecular network of VitD in both COVID-19 and osteoporosis.
As a relatively new approach in drug discovery, network pharmacology and bioinformatics are considered effective methods to clarify the mechanism of drug action37, and have been widely applied to elucidate the interaction between diseases, drugs, and targets, and to screen potential bio-active compounds of drugs35. In this study, we applied these approaches and identified 2660 COVID-19 targets, 5093 osteoporosis targets, and 243 vitamin D targets. In the Venny diagram, we found that COVID-19 shared 855 targets with osteoporosis, implicating COVID-19 and osteoporosis may have shared mechanisms of pathogenesis. We also found that COVID-19, osteoporosis, and VitD shared 42 common targets. Therefore, VitD may mediate a crosstalk between COVID-19 and osteoporosis, which may explain the observations of that VitD insufficiency is closely associated with incidence and clinical manifestation of both COVID-19 and osteoporosis. We further constructed the DTD network of VitD-COVID-19-Osteoporosis based on the PPI network analysis, and 8 targets among the 42 common targets were initially screened as possible important core targets for VitD in the regulation of both COVID-19 and osteoporosis. The GO, KEGG, WikiPathways and Rectome pathway enrichment analyses on the 8 core targets further revealed the detailed mechanisms for VitD regulation of both COVID-19 and osteoporosis. GO analysis revealed that the biological process (BP) regulatory role of VitD mainly acted through cellular response to reactive oxygen species (ROS) and estrogen stimulation. Further screening of the signaling pathway databases, we found that ErbB and MAPK signaling pathway were co-expressed in KEGG and WikiPathways, while the activation pathway of AP-1 transcription factor family in KEGG endocrine resistance pathway is expressed in the immune system of Rectome. These shared pathways also shared common targets, namely ERBB2, EGFR and MAPK8. Among them, EGFR and MAPK8 targets are expressed in the immune system, innate immune system, cytokine signaling in the immune system, GPCR signaling, vesicle-mediated transport and other signaling pathways in the Rectome. Therefore, VitD may exert its regulatory effects mainly through ERBB2, EGFR and MAPK8 mediated 1) activation of the ErbB and MAPK signaling pathways to control the cytokine storm process in COVID-19 patients and to ameliorate pulmonary fibrosis, and 2) activation of osteoimmune mechanisms to regulate the structural integrity of bone. These targets and pathways are relevant for the formation of multiple immune defense mechanisms in COVID-19 and osteoporosis patients. Molecular docking simulations further validated the binding activity between VitD and its target proteins EGFR and MAPK8, reinforcing the importance of the aforementioned targets in the DTD network of VitD-COVID-19-Osteoporosis.
Previous studies have shown that acute SARS-CoV-2 infection often triggers cellular and humoral immune responses38, which can directly or indirectly damage the relevant cells in the respiratory tracts and leads to severe pulmonary fibrosis in COVID-19 patients39. The immune responses in patients with COVID-19 are manifested by marked lymphopenia and elevated serum pro-inflammatory cytokines, as well as a significant infiltration of mesenchymal lymphocytes in lung tissue and excessive activation of T cells in peripheral blood40. Plasma concentrations of IL-1β, IFN-γ, MCP-1 and IP-10 are elevated in COVID-19 patients, which may cause a Th1-type response. In addition, plasma concentrations of Th2 cytokines IL-4, IL-10 and IL-13 were also significantly upregulated in COVID-19 patients41. Thus, the immune response in COVID-19 patients is more inclined to Th1 and Th2 types. In clinically severe cases of COVID-19, an excessive immune response due to an overreaction of the immune system or because the immune system is too weak to control the replication of the virus, causes an inflammatory storm process, also called cytokine storm42, which aggravates the lung damage and causes death. It has been reported that VitD can delay the progression of pulmonary fibrosis43, and can be used as an adjuvant therapy for patients with pulmonary fibrosis44, although the underlying mechanisms are not clear.
The data from our study as described above suggested that VitD may block the immune response of the COVID-19 patients to reduce the pathological process of pulmonary inflammation and fibrosis. As shown in Fig. 9 and Fig. 10, we identified EGFR and ErbB signaling pathways as important targets and signaling pathway of pulmonary fibrosis in patients with COVID-19. In COVID-19 patients, pulmonary fibrosis occurs mainly due to the EGFR-mediated ErbB signaling pathway producing more pro-fibrotic than anti-fibrotic effects. SARS-CoV-2 infection rapidly activates inflammatory T cells and inflammatory monocytes/macrophages in the body, leading to the production of EGFR, AR and TGF-β, and IL-645. These immune cells and inflammatory factors enter the lungs and exert immune damaging effects, leading to severe lung injury or even shock, which may be one of the causes of the SARS-CoV-2 infection induced inflammatory storm46. The ErbB family of receptor tyrosine kinases (RTKs) binds extracellular growth factor ligands to intracellular signaling pathways to regulate various biological responses, including proliferation, differentiation, cell motility, and survival. Ligand binding to four closely related members of this RTK family – EGFR (also known as ErbB-1 or HER1), ErbB-2 (HER2), ErbB-3 (HER3), and ErbB-4 (HER4) - forms homo- and heterodimers of the receptor, activates intrinsic kinase structures, resulting in acid phosphorylation of specific tyrosine residues (pY) in the cytoplasmic tail47. EGFR is characterized by autophosphorylation and phosphorylation of tyrosine residues in the cytoplasmic tail leads to activation of MAPK, JNK and Akt signaling pathways through the ErbB signaling pathway, which leads to inhibition of apoptosis, cell proliferation and migration, activation of inflammatory responses and increased mucus production, leading to lung injury48. EGFR signaling in fibrosis development has a bidirectional regulatory role. Studies have shown that TGF-β1 as a fibrosis inducer can effectively induce the expression of EGFR ligand AR. Silencing of AR using the EGFR-specific small molecule inhibitor gefitinib attenuated the fibrotic effects of TGF-β148. In addition, mice overexpressing the EGFR ligand TGF-α also developed pulmonary fibrosis spontaneously, and similar effects existed for other EGFR ligands. These studies suggest that activation of EGFR signaling is pro-fibrotic49. On the other hand, incidence of interstitial lung disease, a precursor of pulmonary fibrosis, is increased after treatment with tyrosine kinase inhibitors. A similar association with interstitial lung disease has been observed in patients treated with the anti-EGFR monoclonal antibody panitumumab50. The EGFR-specific small molecule inhibitor gefitinib also exacerbated bleomycin-induced pulmonary fibrosis in mice. This suggests that EGFR is anti-fibrotic in specific cases51.
As shown in Fig. 10, we found that MAPK8 and MAPK signaling pathway are also involved in the pathological process of COVID-1947. The involvement of MAPK signaling pathway in COVID-19 is less studied, but it is also closely associated with the major pathological process of pulmonary fibrosis52. As a downstream pathway of the ErbB signaling pathway, MAPK signaling can be activated both by the EGFR-mediated ErbB signaling pathway53 and by factors such as IL-1 and TNF54. In mammals, four main subtype pathways of MAPK signaling pathway exist, and three subtype pathways, p38MAPK, ERK1/2, and JNK, are found to jointly regulate a variety of important cellular physiological and pathological processes such as cell growth, differentiation, stress adaptation to the environment, and inflammatory response. The p38MAPK signaling pathway is phosphorylated in hypoxic environments to regulate inflammatory responses by regulating transcription factor activity and cytokine synthesis55. In addition, the p38MAPK pathway interacts with inflammatory cytokines. Activated p38MAPK can promote the expression and release of various pro-inflammatory cytokines (IL-1β, TNF-α) and induce cytokine storm. Meanwhile, macrophages can be activated by inflammatory cytokines, which then activate the p38MAPK signaling pathway. Other inflammatory cells can also be activated by the p38MAPK signaling pathway, such as the inflammatory aggregation of neutrophils56. These results suggest that the p38MAPK signaling pathway can induce the activation of some inflammatory cells and factors, which eventually cause the generation of inflammatory storms in the lung, leading to lung injury. Studies have shown that patients with idiopathic pulmonary fibrosis have extensive angiogenesis. Angiogenesis is mainly due to a disruption of the balance between stimulating and inhibiting vascular growth factors57. High expression of vascular endothelial growth factor (VEGF) strongly activates ERK1/2, leading to morphological changes in the vasculature, and the growth of neovascularization in the lung will accelerate pulmonary fibrosis58. Finally, as an important branch of the MAPK pathway, the JNK signaling pathway can also lead to lung fibrosis59, and the main mechanisms are that activation of JNK in epithelial cells leads to epithelial-mesenchymal transition (EMT) and cell death, and activation of JNK in lung fibroblasts leads to myofibroblast phenotype60. Therefore, MAPK signaling pathway mediates inflammatory storm, lung angiogenesis, fibroblast to myofibroblast conversion and pulmonary fibrosis development in COVID-19 patients. Inhibition of the ErbB and MAPK signaling pathways by VitD reduces TGF-β signaling and inhibits EGFR ligand-dependent phosphorylation and attenuates the cytokine storm generated by the immune response to SARS-CoV-2 infection61 .
There exists an intrinsic connection between the skeletal system and the immune system, which is termed osteoimmunity, where cytokines and signaling pathways are the bridges between the two intrinsic connection58. Therefore, the series of immune responses occurring in COVID-19 patients would also affect the skeletal system through osteoimmune responses, thus inducing osteoporosis or aggravating the condition of osteoporosis patients. The key immune cells of osteoimmunity are T cells, B cells, dendritic cells, and bone marrow macrophages. The RANKL/RANK/OPG regulatory system is the main signaling pathway of the bone immune response. The main cytokines that favor bone formation are IL-1, IL-6, IL-17, and TNF, and the main cytokines that aggravate bone destruction are IL-4, IL-13, and INF-γ62. Our data analysis found that SARS-CoV-2 infection was able to activate T cells and inflammatory monocytes/macrophages, led to the production of EGFR, AR and TGF-β, IL-6, where the MAPK signaling pathway is also a downstream pathway belonging to the RANKL/RANK/OPG regulatory system63. EGFR not only regulates pulmonary fibrosis, but also regulates bone structural integrity64. It was found that EGFR inhibited the expression of osteoblast (OB) transcription factors Runx2 and Osterix, thereby suppressing osteoblast differentiation65. EGFR stimulates OB proliferation and inhibits their differentiation by inhibiting the IGF-1R / mTOR pathway through ERK1/ 2-dependent upregulation of IGFBP-366. OB derived from multipotent bone marrow mesenchymal stem cells (MSC) plays an important role in bone structural integrity64, but MSCs have a short survival time and require constant replenishment65. Insufficient MSC leads to low bone formation activity, which increases the risk of osteoporosis58. Chandra et al. found that the activation of EGFR signaling increased the number of MSC and facilitated the promotion of OB formation59. The main mechanism of this process is that EGFR promotes the proliferation and survival of osteogenic progenitor cells by increasing the expression of early growth response factor 2 (EGR2). EGR2 promotes the proliferation and survival of OB by increasing the anti-apoptotic protein MCL1 and decreasing the apoptosis of OB59. Some of the above evidence suggest that the main role of EGFR is to promote bone formation. The MAPK signaling pathway is a downstream of the bone immune RANKL/RANK/OPG regulatory system and is critical in controlling OB differentiation and skeletogenesis67. The basic composition of the MAPK pathway is a three-tier kinase response pattern that includes MAPK kinase kinase (MAP kinase kinase kinase, MKKK), MAPK kinase kinase (MKK), and MAPK, which can be activated sequentially and together regulate a variety of important cellular physiological/pathological processes such as cell growth, differentiation, stress adaptation to the environment, and inflammatory responses. And p38MAPK is primarily involved in OB differentiation, skeletogenesis, and skeletogenesis through a series of kinase reactions in OB differentiation, Osteoclast (OC) formation and apoptosis[64]. Studies have shown that p38MAPK is an important positive regulator of OB function and bone formation in vivo68. ERK in the p38MAPK transduction pathway plays an important role in OB proliferation, adhesion, extension, migration and integration. ERK1/2 positively regulates OB differentiation and inhibits chondrocyte differentiation69. ERK also affects RANKL to cause OC activation, leading to enhanced osteolysis70. Finally, JNK signaling is also involved in OB differentiation, OC formation and apoptosis71. Multiple factors stimulate intracellular signaling aggregation in the MAPK pathway, which affects OB and OC proliferation, differentiation, and apoptosis70. It has been shown that the synergistic presence of VitD with p38MAPK contributes to skeletal muscle growth and regeneration. Therefore, our data support the lotion that VitD may improve osteoporosis by binding to MAPK signaling pathway mediated osteoimmunity.
In summary, in this study we identified and characterized 42 common targets of VitD on both COVID-19 and osteoporosis and 8 core targets in the DTD network of VitD-COVID-19-osteoporosis. These VitD targets involved in the ErbB and MAPK signaling pathways are critical for the regulation of lung fibrosis, bone structural integrity, and systematic immune responses. These findings provided novel mechanistic insights into the functional roles and molecular network of VitD in both COVID-19 and osteoporosis. VitD may be used as a marker of poor prognosis or a possible risk factor for both COVID-19 and osteoporosis and supplementation of VitD may have beneficial effects for prevention and treatment of these devastating diseases.