We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

The COVID-19 pandemic: an increased risk of rheumatoid arthritis

    Archana Tripathy

    Disease Biology Laboratory, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) deemed to be University, Bhubaneswar, Odisha, 751024, India

    ,
    Nitish Swain

    Disease Biology Laboratory, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) deemed to be University, Bhubaneswar, Odisha, 751024, India

    &
    Bhawna Gupta

    *Author for correspondence: Tel.: +91 993 746 6890;

    E-mail Address: bhawna.gupta@kiitbiotech.ac.in

    Disease Biology Laboratory, School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) deemed to be University, Bhubaneswar, Odisha, 751024, India

    Published Online:https://doi.org/10.2217/fvl-2020-0393

    Abstract

    COVID-19 is a respiratory infection similar to viral pneumonia and is caused by SARS-CoV-2. Chloroquine and hydroxychloroquine make up the major part of the treatment regimen for the management of COVID-19 infections, which are also commonly used in treatment of patients with malaria as well as autoimmune diseases like rheumatoid arthritis (RA). In this review, we analyzed the scientific evidences pertaining to any possible association of SARS-CoV-2 infection with RA. We thus believe that people predisposed to RA carry a higher infection risk than the general population both due to the iatrogenic effects of the RA related drug therapy. Thus COVID-19 pandemic may bring a higher risk of health emergency in complex diseases such as RA.

    The year 2020 unfortunately has seen a dark beginning with the spread of a novel coronavirus pandemic COVID-19. Alhough the disease initiated from the Wuhan province of China, it has now spread to more than 200 countries worldwide. As of 21 June 2021 COVID-19 has affected more than 177 million people worldwide with above 3.8 million deaths. The countries have locked down international borders, asked residents to stay home and work from home with only essential services running. Amidst the uncertainty people suffer from economical downfall, anxiety and stress while COVID-19 epidemiology is rapidly evolving with an increasing number of cases on a daily basis.

    Although a number of coronaviruses of the family Coronaviridae have been known to infect humans [1] yet the infections have been mild and resulted in common cold and flu-like symptoms. New and pathogenic strains of these viruses (MERS-CoV, SARS-CoV) have emerged with high fatality rates resulting in acute respiratory distress syndrome, reduced lung function, arrhythmia and eventually death. COVID-19 is known to be caused by a mutated strain of SARS-CoV that is reported to be enveloped with a positive-sense ssRNA genome and hence named SARS-CoV-2. The human angiotensin-converting enzyme 2 (ACE2) is identified as the host cell-surface receptor for the envelope spike glycoprotein of SARS-CoV-2, facilitating its entry and infection in the host cell [2]. The ACE2 is a cell membrane receptor expressed on a number of different types of cells including the cells of the GI tract, blood vessels, lung AT2 alveolar epithelial cells and others [3]. The interaction of ACE2 with SARS-CoV-2 spike protein results in ACE2 downregulation, an increased production of angiotensin II, a subsequent activation of type 1a angiotensin II receptor (AT1RA) that increases pulmonary vascular permeability thereby increasing lung damage [4,5]. COVID-19 thus results in a chronic condition and hence a pandemic situation has arisen. The COVID-19 is also detrimental to the debilitating population with autoimmune rheumatic diseases that are either at the higher risks of infection leading to disease severity or may suffer due to the induced effects of the immune-suppressive agents like the disease-modifying antirheumatic drugs.

    Rheumatoid arthritis (RA) is one of the most prevalent and debilitating autoimmune diseases characterized by the inflammation of the synovium [6,7]. The patients suffer from acute pain and swelling of joints such that they are unable to move, walk, hold objects and thus become dependent on others for their daily activities. Although etiology of RA is unknown yet it is linked to several genetic and environmental factors [8,9]. This disease condition is marked by presence of a dysfunctional immune system that recognizes self antigens as foreign. The preclinical phase is said to be characterized by the generation of auto antibodies and the clinical phase is attained when the body reacts to these auto-antibodies leading to inflammation [8]. The inflammatory condition is driven by the cytokines that are upregulated during the disease condition. Th1cells producing IFN-γ and Th17 cells producing IL-17 are the key regulators for disease progression [10,11]. The release of granzyme and perforin by the activated CD8+ T cells is also known to aggravate the disease condition [12]. The similarities in the cytokine profile, lymphocyte population characteristics and inflammatory mediators interestingly present an intricate relationship between COVID-19 and RA. We thus have tried to find potential reasons of co-existence of COVID-19 and RA as well as the repercussions of COVID-19 pandemic in the increase in population of RA patients (Figure 1) worldwide.

    Figure 1. SARS-CoV-2 infection can potentiate initiation, progression and perpetuation of rheumatoid arthritis using parallel mechanisms.

    RA: Rheumatoid arthritis.

    Methodology

    A nonsystematic review for published literature was carried out in PubMed and Google Scholar databases. Articles published in 2020 having terms “SARS-CoV-2” OR “COVID-19” OR “Rheumatoid Arthritis” in the title were considered.

    In this study the inclusion of articles was done on the model proposed by the User’s Guide to Medical Literature (JAMA Network).

    Excessive production of angiotensin II by SARS-CoV-2 infection

    SARS-CoV-2, a positive-sense ssRNA virus, has the genome size of 26–32 kb, a crown-like appearance under an electron microscope with a confirmed presence of spike glycoprotein on the envelope [1]. Two-thirds of the genome open reading frame toward 5’ terminal encodes replicase transcriptase complex and rest of the genome encodes the structural proteins including spike (S), envelope (E), nucleocapsid (N) and membrane (M) proteins [13]. The entry of corona virus into the host cells is a result of attachment of spike (S) protein to host cellular receptors called ACE2 [4,14,15] resulting in an increased expression of serum angiotensin II [5]. Angiotensin II is recognized as a powerful pro-inflammatory mediator that acts through stimulation of angiotensin II receptor (AT1R), present on the cell surface of lymphocytes, macrophages and other immune cells [16,17]. The presence of angiotensin II receptor and its elevated expression in the synovium of RA patients leads to synovial hyperplasia [18,19]. Kawakami et al. showed that the cytokines like IL-1 regulates the expression of both the angiotensin II receptors, AT1R and AT2R, in articular chondrocytes in RA patients [20]. AT1R antagonist, losartan reduces the level of C-reactive protein (CRP) and erythrocyte sedimentation rate of RA patients [21]. An in vitro study showed that the losartan treatment suppressed TNF-α production in synovium of inflamed joints of RA patients [19]. Excessive angiotensin II concentration initiates inflammatory response by stimulating the production of VEGF and prostaglandins as well as increasing vascular permeability [22]. Angiotensin II as well activates monocytes and upregulates the expression of TNF-α, IL-6 and IL-8, which are the potent activators of neutrophils [23]. In addition, it also increases CRP transcripts and protein in macrophages via NF-κB activation and angiotensin receptor mediated reactive oxygen species (ROS) production [24]. angiotensin II can directly enhance the production of ROS in leucocytes and stimulate the activation of lymphocytes [25]. Therefore, angiotensin II signaling through AT1R subsequently produces pro-inflammatory cytokines and chemokines by peripheral blood cells which contributes to the migration of immune cells to the site of injury and multiplies the inflammatory response (Figure 2) [26]. Above reports may potentially explain that the genetically predisposed individuals for RA if infected with SARS-CoV-2 may suffer from the disease initiation, while the already existing RA patients may see a surge in the disease severity. Also the excessive presence of angiotensin II due to the infection of SARS-CoV-2 will stimulate AT1R and AT2R, which may aggravate symptoms and severity of RA.

    Figure 2. The inflammatory roles of angiotensin II on innate and adaptive immune response.

    Ang II via AT1R signaling in immune cells contributes to the secretion of pro-inflammatory mediators and enhances inflammatory responses by activating innate and adaptive immune cells.

    Ang II: Angiotensin II; DC: Dendritic cell; NK: Natural killer.

    Cellular immune response by SARS-CoV-2 infection

    Innate immune cells are important target cells of angiotensin II and express both AT1R and AT2R, which have substantial roles in production of metalloproteinase and promoting vascular inflammation. Xu et al. recently performed peripheral blood cell profiling of COVID-19 patients by flow cytometry [27]. They found the number of CD4+ as well as CD8+ T cells declined significantly in SARS-CoV-2 infected patients, but the cells were highly activated, which was confirmed by the presence of high proportions of HLA-DR (CD4: 3.47%) and CD38 (CD8: 39.4%) double positive populations. Increased concentration of pro-inflammatory CCR6+ Th17 subpopulation of CD4+ T cells was also identified [27]. Additionally, CD8+ T cells were found with higher concentration of cytotoxic granules wherein 64.2% cells were granulysin positive, 31.6% cells were perforin positive while 30.5% cells were both perforin and granulysin positive [27]. This data imply that over activation of T cells, increased percentage of Th17 cells and high cytotoxic activity of CD8+ T cells all together leads to severe immune injury and may increase the chance of incidence of T lymphocyte mediated diseases like RA.

    Angiotensin II acts as a co-stimulatory signal for activation, differentiation and proliferation of T cells via AT1R [28,29]. Angiotensin II induces Th1 differentiation by increasing the expression of IFN-γ in addition with reduction of IL-4 [29]. It also stimulates the production of two prominent inflammatory cytokines, IFN-γ and TNF-α, in T cells (Figure 2) [30]. Commercially available AT1R blockers like olmesartan, telmisartan and candesartan have also been shown to inhibit antigen specific Th1 and Th2 immune responses [31]. Wu et al. demonstrated that angiotensin II also induces Th17 responses [32]. Th1 and Th17 differentiation by angiotensin II thus suggests progression of RA upon SARS-CoV-2 infection.

    Involvement of Toll-like receptor during coronavirus infection

    TLR7, an endosomal receptor for ssRNA, is present on the endosomal membrane [33,34]. It can differentiate self RNA from nonself viral ssRNAs due to the presence of higher number of modified nucleotide bases (m5C, m5U, m6A and s2U) in cellular RNA [35]. TLR7 is predominantly expressed by RA synovial macrophages and by RA synovial fibroblasts cells [36]. Expression of TLR7 is also upregulated in RA monocytes and shows a strong positive correlation with TNF-α levels as well as DAS28 [36]. It has been reported that the SARS-CoV infection induces upregulation of TLR7 in monocytes (THP1 cells) however the downstream signaling mechanism is not clear till date [37]. TLR7 signaling promotes the transcription of cytokines involved in Th17 cell differentiation as well as inhibits the TGF-β signaling [38,39]. TGF-β is widely recognized for its role in reducing inflammation and maintaining pulmonary homeostasis by inhibiting both Th2 and Th1 responses [40,41]. It thus becomes imperative to understand if the increased TLR7 in RA patients can efficiently recognize ssRNA of SARS-CoV-2 and initiates the TLR7 mediated inflammatory response further potentiating disease severity.

    Cytokine storm in COVID-19

    COVID-19 is considered as cytokine storm syndrome because of robust increase in several cytokines and chemokines in addition to the cytolytic enzymes (Figure 3). Patients with novel COVID-19 show elevated serum levels of IFN-γ, TNF-α, IFN-γ-induced protein 10 (IP 10), monocyte chemo attractant protein-1 (MCP-1), macrophage inflammatory proteins (MIP) which belongs to the family of chemotactic cytokines known as chemokines, granulocyte-colony stimulating factor, granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-1β, IL-2, IL-7, IL-8, IL-9, IL-17 and others [42,43]. Cytolytic enzymes like perforin and granulysin are also increasingly secreted by CD8+ T cells of COVID-19 patients [27].

    Figure 3. Schematic diagram represents the comparison of inflammatory responses by a healthy individual with those by a rheumatoid arthritic patient when infected with SARS-CoV-2.

    Cytokine storm causes uncontrolled immune response cycle in rheumatoid arthritic patients and begins to destroy healthy/self-cells. But in case of healthy individuals infected with SARS-CoV-2, they may recover with mild symptoms after a series of inflammatory reactions by innate and adaptive immune cells.

    RA: Rheumatoid arthritis.

    Among these, several cytokines are involved in inducing Th1 and Th17 type responses (Figure 3). TNF-α and IL-1β both induce Th17 lineage differentiation of naive CD4+ T cells and promote vascular permeability and leakage [44]. TNF-α is concomitantly highly expressed by both Th1 and Th17 cells [45]. Th17 cells secrete IL-17 and GM-CSF [46,47]. IFN-γ and GM-CSF secretions are also associated with Th1 cells in humans [48,49]. IL-21 is required for Th17 cell maintenance in a STAT-3 dependent manner [50]. IL-17 has broad pro-inflammatory effects on induction of cytokines like TNF-α, GM-CSF, IL-1β and IL-6 (induce systematic inflammatory response); chemokines like IP10, MIP2A, IL-8, MIP3A (attract and recruit immune infiltrates) and matrix metalloproteinases (MMP) (participate in tissue damage and remodeling) [32].

    RA is known to be a Th1/Th17 disorder wherein naive CD4+ T cells increasingly differentiate into Th1 and or the Th17 lineage. Several pro-inflammatory mediators known to be responsible for an increased differentiation of naive CD4+ T cells to Th1/Th17 phenotype is found to be increased in COVID-19 patients. Taken together, we thus propose that cytokine storm developed by Th1 and Th17 type responses during SARS-CoV-2 infection may increase the incidence of RA and trigger a violent attack by the immune system thereby making it difficult for the patient to recover from the SARS-CoV-2 infection.

    Pathways associated with SARS-CoV-2 infection

    Several cytokines including IFN-γ, GM-CSF, IL-6 and IL-15 involved in RA pathogenesis activate janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway [51]. STAT-3 is constitutively activated in RA patients [51]. Due to excessive presence of IFN-γ and GM-CSF in the serum of a COVID-19 patient [27], these can activate JAK/STAT pathway and lead to the upregulation of several interferon stimulated genes [52] which can elevate the amount of cytokines having the ability to attack self cells and induce inflammation in RA. JAK/STAT mediated activation of immune cells by SARS-CoV-2 infection may thus contribute in severity and progression of RA. It is already known that angiotensin II level is high in COVID-19 patients and its signaling through AT1R leads to activation of NF-κB with subsequent synthesis of pro-inflammatory cytokines, chemokines, cytolytic enzymes and cell adhesion molecules [53,54]. They contribute to the migration of immune cells to the site of infection and amplify the inflammatory responses. MAPK pathways comprises of the p38 mitogen activated protein kinases (MAPK), extracellular signal regulated kinase (ERK) and janus kinase (JNK) pathways [55]. During SARS-CoV and MERS-CoV infections, the signals are transmitted by a series of protein kinase cascades of MAPK pathways, with phosphorylation at Tyr and Thr residues [56]. During coronavirus infection, viral protein triggers the activation of MAPK pathways, which in turn regulate the production of variety of cytokines [57]. All these intermediates of transcription pathway associated with SARS-CoV infection collectively produce inflammatory cytokines, which can increase the incidence of infection in RA and may progressively increase the disease severity in RA patients.

    Other related consequences of SARS-CoV-2 infection in RA

    Changes in clinical parameters

    RA is characterized by perturbations in the levels of different clinical parameters like erythrocyte sedimentation rate (ESR), CRP, anti-CCP and rheumatoid factor. Recent reports of COVID-19 suggest significant upregulation in the levels of ESR and CRP post infection [58]. Studies with SARS-CoV have revealed an increase in CRP levels in the affected population, whereas in MERS-CoV rise in both CRP and ESR levels have been reported [59–61]. Respiratory viral infection studies have also revealed higher expression of CRP and ESR [62,63]. Thus, excessive secretion of angiotensin II post infection by SARS-CoV-2 increases two specific disease parameters of RA namely ESR and CRP [21].

    Genetic factors

    RA is classified as HLA II (MHC II) associated disease and HLA-DRB1 alleles are the most relevant genetic factors for disease susceptibility [64]. The antigen presentation of MERS-CoV infection mainly depends on MHC II molecules [65], whereas SARS-CoV depends on both MHC I and MHC II molecules [66]. As the mechanism of antigen presentation of SARS-CoV-2 is still unclear, we can predict a genetic predisposition toward COVID-19 in RA.

    Clinical studies

    The clinical studies that have taken place in the past year on COVID-19 and RA are mostly case studies. The case studies have mostly focused on the levels of markers of RA in people who have encountered COVID-19. Increase in ESR, Anti-CCP, IL-6 and CRP has been reported in patients who had been affected by COVID-19 with history of RA [67–69]. These signature clinical parameters have also been reported to increase post COVID-19 infection and patients have shown symptoms of early onset of RA. Presence of anti-PAD2, anti-PAD4 along with Sjogren syndrome related antibodies have been reported too [69]. Patients with remising RA were seen to have a flare in disease activity post infection with COVID-19 [70].

    Effects of biologic disease-modifying anti-rheumatic drugs

    Biologic treatments have been in use for the treatment of RA due to their less side effects as compared with conventional disease-modifying anti-rheumatic drugs (DMARDs). The mostly used biologic disease modifying anti-rheumatic drugs (bDMARDs) are tocilizumab, eternacept, anakinra and barcitinib. In case of RA these bDMARDs act as IL-6, TNF-α, JAK and IL-1 blockers, respectively and bring down the production of cytokines though rendering the patient immunosuppressed [71]. In case of COVID-19 the initiation of cytokine release syndrome takes place in severe COVID-19 infection. These bDMARDs have been introduced as a supplementary treatment regimen along with the use of antivirals. When administered these bDMARDs block their specific targets and help in relieving the cytokine load on the infected patient thereby partially eliminating the risk of cytokine release syndrome [72,73]. The main drawback of this therapy is the state of immunosuppression that occurs in the patient which may lead to several opportunistic infections. Research and case studies have proposed the supplemented use of these biologic DMARDs in combination with antivirals and with proper timely assessment of the patient to look for the opportunistic infections [72,74].

    Recurrence of COVID-19 & similarities with recurrence of RA

    The sequel or recurrence of COVID-19 has mostly been seen in older patients and can be attributed to numerous factors. The patient post recovery has a very poor physical health, poor resistance and impaired physical functions [75]. The poor immune function after the duration of treatment also increases the chances of re-infection. The treatment regimen includes use of glucocorticoids which inhibit the body immune function and increase the risk of secondary infection [76]. Co-morbidities associated with the patient are also a major contributing factor toward the recurrence of COVID-19. The recurrence of COVID-19 is similar to the relapse of RA in the aspects of age, physical health status and physical function. RA relapse mostly occurs in patients who have had the disease for long periods of time and with old age the body being immune suppressed acquires other infections. The associated co-morbidities like Type 2 diabetes mellitus (T2DM), interstitial lung disease (ILD) and osteoporosis also trigger the relapse of RA [77].

    Diagnostic findings

    As of now the gold standard for diagnosis of COVID-19 is the real-time (RT)-PCR test. The lack of diagnostic findings in both diseases are due to their complicated natures. COVID-19 being a viral infection has high possibilities of mutations and in case of RA the etiology is still unknown [78]. The specific reason for the onset of RA is yet to be elucidated. Current test for RA diagnosis is based on the levels of various inflammatory markers like complete blood count, CRP and serum glutamic pyruvic transaminase and these markers are also monitored in COVID-19 to check for the onset of cytokine release syndrome [79]. To increase the diagnostic findings in case of both the diseases we have to focus our research on the etiology and with this find out novel markers for better treatment of both the diseases.

    Preventive medication

    The use of chloroquine derivatives such as hydroxychloroquine was started due to its antiviral and anti-inflammatory properties but the major pushing force was its availability and the inexpensive nature of the drug. Hydroxychloroquine modulates the infection by acting on the viral entry mechanism. It has been shown to make the ACE2 receptor malfunction and hence treat the patients [80].

    But the use of hydroxychloroquine showed lack of preventive role because this drug failed to act on the viral replication. It was seen that in comparison to other antivirals, hydroxychloroquine did not prove beneficial [81]. As the treatment required high doses of hydroxychloroquine certain side effects including gastrointestinal symptoms, pruritus and dermatological changes which have further discouraged its use for treatment of COVID-19 [82].

    Conclusion

    COVID-19 has presented itself as the greatest global crisis of this millennium and has affected almost all the countries in the world. Research on the pathophysiology of COVID-19 has brought into light different aspects of this novel disease. The increased levels of angiotensin II which is one of the key characteristic of the disease has been shown to up regulate the levels of pro-inflammatory cytokines like TNF-α, IL-6 and IL-8 which are significantly increased in case of RA [83,84]. Angiotensin has also been correlated with the production of IFN-γ which is a key cytokine in case of RA. The increase in the level of CRP can be seen as a link to the potential occurrence of RA in a COVID-19 patient [24,25,85]. High ROS production and activation of NF-κB are some of the other factors that may govern the incidence of RA [25]. The highly activated lymphocytic population and Th1/Th17 responses post infection with SARS-CoV-2 can be seen as a key factor for the occurrence of RA in COVID-19 patients [86]. The activation of JAK/STAT, NF-κB and MAPK pathways during the SARS-CoV infection resulting in the synthesis of several inflammatory mediators might just trigger the genetic factors associated with RA or might lead to an aggravated immune response [27]. The suppressed immune system in case of RA, due to the use of conventional and biologic DMARDs may present complications if infected with SARS-CoV-2. This may prolong the onset of cytokine release syndrome but may open up the victim to other opportunistic infections that might increase morbidity. There have been studies where the relation between other respiratory viral infections and RA has been shown. Thus, we propose that RA patients might be predisposed to disease severity post SARS-CoV-2 infection suggesting that severe RA and COVID-19 might coexist and the genetically predisposed individuals to RA may have a high risk to develop RA if they get infected with SARS-CoV-2.

    Future perspective

    For a better understanding of SARS-CoV-2 we need a series of strategic research especially with respect to RA. RA being a complicated auto-immune disorder will only add to the cytokine burden in a patient with SARS-CoV-2 infection. The information obtained while analyzing the case studies with RA patients suffering from SARS-CoV-2 must be made available to clinicians worldwide for the better management of the disorder.

    Executive summary
    • In this review we have tried to find potential reasons of co-existence of COVID-19 and rheumatoid arthritis (RA) as well as the repercussions of COVID-19 pandemic in the population of RA patients worldwide.

    Excessive production of angiotensin II by SARS-CoV-2 infection

    • The entry of SARS-CoV-2 is a result of attachment to host cellular receptor angiotensin-converting enzyme 2. The presence of angiotensin II in RA initiates inflammatory response by activation of monocytes and upregulates the expression of TNF-α, IL-6 and IL-8. Increase in C-reactive protein (CRP) and reactive oxygen species also help in activation of lymphocytes. Therefore, Angiotensin II signaling through AT1R subsequently leads to the production of chemokines and pro-inflammatory cytokines and contributes to the activation and migration of immune cells to the target site and elicits the immune response.

    Cellular immune response by SARS-CoV-2 infection

    • The increase in the cytotoxic activity of the CD8+ T cells has been seen in SARS-CoV-2. The increase in Th17 fate of immune cells might increase the chances of a Th17-mediated disease like RA. Angiotensin II has also been found to act as a co-stimulatory signal for activation, differentiation and proliferation of T cells via AT1R, leading to production of IFN-γ and TNF-α. This Th1 and Th17 fate of the T cells suggest onset or progression of RA upon SARS-CoV-2 infection.

    Involvement of Toll-like receptor during coronavirus infection

    • TLR7 is an endosomal receptor present in immune cells and recognizes ssRNA. The recognition of SARS-CoV-2 genetic material by TLR7 might lead to a Th17-driven immune response, RA being a Th17-driven disorder might become more aggravated upon infection with SARS-CoV-2. The downstream events occurring must be studied carefully to know he interaction that occurs between SARS-CoV-2 viral genome and if that can lead to an aggravated form of RA.

    Cytokine storm in COVID-19

    • Cytokine storm is seen in patients with very severe cases of COVID19, robust increase in levels of IFN-γ, TNF-α, macrophage inflammatory proteins, granulocyte-macrophage colony-stimulating factor, IL-1β, IL-2, IL-7, IL-8, IL-9, IL-17, perforin and granulysin has been reported. These inflammatory molecules play a major role in the disease progression in a patient suffering from RA. The onset of such cytokine storm might lead to the onset or progression of RA.

    Pathways associated with SARS-CoV-2 infection

    • Cytokines like IFN-γ and granulocyte-macrophage colony-stimulating factor which are highly activated in RA are potent activators of JAK/STAT pathway. COVID-19 also presents expression of these cytokines in large quantities, this may lead to the activation of the JAK/STAT pathway and induce inflammation. The AT1R signaling leads to activation of NF-κB which results in the synthesis of numerous inflammatory molecules.

    Other related consequences of SARS-CoV-2 infection in RA

    • RA is characterized by perturbations in the levels of different clinical parameters like ESR, CRP, Anti-CCP and rheumatoid factor. Recent reports of COVID-19 suggest significant up regulation in the levels of ESR and CRP postinfection. Genetic factors like HLA II (MHC II) associated disease and HLA-DRB1 alleles are the most relevant genetic factors for disease susceptibility. The antigen presentation of MERS-CoV infection mainly depends on MHC II molecules, whereas SARS-CoV depends on both MHC I and MHC II molecules. As the mechanism of antigen presentation of SARS-CoV-2 is still unclear, we can predict a genetic predisposition toward COVID-19 in RA. Biologic DMARDs have been introduced as a supplementary treatment regimen along with the use of antivirals. When administered these bDMARDs block their specific targets and help in relieving the cytokine load on the infected patient thereby partially eliminating the risk of cytokine release syndrome.

    Future directions

    • Research needs to focus on large cohorts rather than individual case studies. This would enable the researchers to find out the key aspects of COVID-19 and RA relationship.

    Author contributions

    A Tripathy, N Swain, B Gupta designed the manuscript; A Tripathy, N Swain drafted the manuscript; A Tripathy, N Swain, B Gupta revised and approved the manuscript.

    Financial & competing interests disclosure

    A Tripathy is thankful to Department of Science and Technology for Senior Research Fellowship (ref no. DST/INSPIRE/03/2014/000394). N Swain expresses his gratitude to University Grants Commission for Junior Research Fellowship (ref no. 420/[CSIR-UGC NET DEC. 2017]). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Su S, Wong G, Shi W et al. Epidemiology, genetic recombination, and pathogenesis of coronaviruses. Trends Microbiol. 24(6), 490–502 (2016).
    • 2. Hoffmann M, Kleine-Weber H, Schroeder S et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 181(2), 271–280 (2020). •• Sheds light on the mechanism of entry of SARS-CoV-2 into the host cell leading to infection and subsequent diagnosis of COVID-19.
    • 3. The Human Protein Atlas. SARS-CoV-2 related proteins. https://www.proteinatlas.org/humanproteome/sars-cov-2
    • 4. Zhao Y, Zhao Z, Wang Y, Zhou Y, Ma Y, Zuo W. Single-cell RNA expression profiling of ACE2, thereceptor of SARS-CoV-2. Am. J. Respir. Crit. Care Med. 202(5), 756–759 (2020). • Reports the RNA expression profile of angiotensin-converting enzyme2 in human lung cells.
    • 5. Zou Z, Yan Y, Shu Y et al. Angiotensin-converting enzyme 2 protects from lethal avian influenza A H5N1 infections. Nat. Commun. 5(1), 1–7 (2014).
    • 6. Firestein GS. Evolving concepts of rheumatoid arthritis. Nature 423(6937), 356–361 (2003).
    • 7. Tripathy A, Khanna S, Padhan P, Smita S, Raghav S, Gupta B. Direct recognition of LPS drive TLR4 expressing CD8+ T cell activation in patients with rheumatoid arthritis. Sci. Rep. 7(1), 1–10 (2017).
    • 8. Firestein GS, Mcinnes IB. Immunopathogenesis of rheumatoid arthritis. Immunity 46(2), 183–196 (2017).
    • 9. Joo YB, Lim Y-H, Kim K-J, Park K-S, Park Y-J. Respiratory viral infections and the risk of rheumatoid arthritis. Arthritis Res. Ther. 21(1), 199 (2019). •• Shows relationship between viral infection and how it affects the onset of rheumatoid arthritis.
    • 10. Vukmanovic-Stejic M, Vyas B, Gorak-Stolinska P, Noble A, Kemeny DM. Human Tc1 and Tc2/Tc0 CD8 T-cell clones display distinct cell surface and functional phenotypes. Blood 95(1), 231–240 (2000).
    • 11. Alzabin S, Williams RO. Effector T cells in rheumatoid arthritis: lessons from animal models. FEBS Lett. 585(23), 3649–3659 (2011).
    • 12. Carvalheiro H, Duarte C, Silva-Cardoso S, Da Silva JA, Souto-Carneiro MM. CD8+ T cell profiles in patients with rheumatoid arthritis and their relationship to disease activity. Arthritis Rheumatol. 67(2), 363–371 (2015).
    • 13. Li X, Geng M, Peng Y, Meng L, Lu S. Molecular immune pathogenesis and diagnosis of COVID-19. J. Pharm. Anal. 10(2), 102–108 (2020). •• Discusses current knowledge of molecular immune pathogenesis, diagnosis and treatment of COVID-19.
    • 14. De Wit E, Van Doremalen N, Falzarano D, Munster VJ. SARS and MERS: recent insights into emerging coronaviruses. Nat. Rev. Microbiol. 14(8), 523 (2016).
    • 15. Fukao T, Fukuda Y, Kiga K et al. An evolutionarily conserved mechanism for microRNA-223 expression revealed by microRNA gene profiling. Cell 129(3), 617–631 (2007).
    • 16. Nataraj C, Oliverio MI, Mannon RB et al. Angiotensin II regulates cellular immune responses through a calcineurin-dependent pathway. J. Clin. Investig. 104(12), 1693–1701 (1999).
    • 17. Marchesi C, Paradis P, Schiffrin EL. Role of the renin–angiotensin system in vascular inflammation. Trends Pharmacol. Sci. 29(7), 367–374 (2008).
    • 18. Walsh DA, Suzuki T, Knock GA, Blake DR, Polak JM, Wharton J. AT1 receptor characteristics of angiotensin analogue binding in human synovium. Br. J. Pharmacol. 112(2), 435–442 (1994).
    • 19. Price A, Lockhart J, Ferrell W, Gsell W, Mclean S, Sturrock R. Angiotensin II Type 1 receptor as a novel therapeutic target in rheumatoid arthritis: in vivo analyses in rodent models of arthritis and ex vivo analyses in human inflammatory synovitis. Arthritis Rheum. 56(2), 441–447 (2007).
    • 20. Kawakami Y, Matsuo K, Murata M et al. Expression of angiotensin II receptor-1 in human articular chondrocytes. Arthritis 2012, 2012 (2012).
    • 21. Perry M, Chee M, Ferrell WR, Lockhart J, Sturrock R. Angiotensin receptor blockers reduce erythrocyte sedimentation rate levels in patients with rheumatoid arthritis. Ann. Rheum. Dis. 67(11), 1646–1647 (2008).
    • 22. Williams B, Baker AQ, Gallacher B, Lodwick D. Angiotensin II increases vascular permeability factor gene expression by human vascular smooth muscle cells. Hypertension 25(5), 913–917 (1995).
    • 23. Nobuhiko A, Suganuma E, Babaev VR et al. Angiotensin II amplifies macrophage-driven atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 24(11), 2143–2148 (2004).
    • 24. Han C, Liu J, Liu X, Li M. Angiotensin II induces C-reactive protein expression through ERK1/2 and JNK signaling in human aortic endothelial cells. Atherosclerosis 212(1), 206–212 (2010).
    • 25. Dandona P, Kumar V, Aljada A et al. Angiotensin II receptor blocker valsartan suppresses reactive oxygen species generation in leukocytes, nuclear factor-κB, in mononuclear cells of normal subjects: evidence of an antiinflammatory action. J. Clin. Endocrinol. Metab. 88(9), 4496–4501 (2003).
    • 26. Chang Y, Wei W. Angiotensin II in inflammation, immunity and rheumatoid arthritis. Clin. Exp. Immunol. 179(2), 137–145 (2015).
    • 27. Xu Z, Shi L, Wang Y et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 8(4), 420–422 (2020).
    • 28. Goldszmid RS, Caspar P, Rivollier A et al. NK cell-derived interferon-γ orchestrates cellular dynamics and the differentiation of monocytes into dendritic cells at the site of infection. Immunity 36(6), 1047–1059 (2012).
    • 29. Hoch NE, Guzik TJ, Chen W et al. Regulation of T-cell function by endogenously produced angiotensin II. Am. J. Physiol. Regul. Integr. Comp. 296(2), R208–R216 (2009).
    • 30. Shao J, Nangaku M, Miyata T et al. Imbalance of T-cell subsets in angiotensin II–infused hypertensive rats with kidney injury. Hypertension 42(1), 31–38 (2003).
    • 31. Mazzolai L, Duchosal MA, Korber M et al. Endogenous angiotensin II induces atherosclerotic plaque vulnerability and elicits a Th1 response in ApoE−/− mice. Hypertension 44(3), 277–282 (2004).
    • 32. Wu D, Yang XO. TH17 responses in cytokine storm of COVID-19: an emerging target of JAK2 inhibitor Fedratinib. J. Microbiol. Immunol. Infect. 53(3), 368–370 (2020).
    • 33. Heil F, Hemmi H, Hochrein H et al. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science 303(5663), 1526–1529 (2004).
    • 34. Lund JM, Alexopoulou L, Sato A et al. Recognition of single-stranded RNA viruses by Toll-like receptor 7. Proc. Natl. Acad. Sci. USA 101(15), 5598–5603 (2004).
    • 35. Karikó K, Buckstein M, Ni H, Weissman D. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity 23(2), 165–175 (2005).
    • 36. Chamberlain ND, Kim S-J, Vila OM et al. Ligation of TLR7 by rheumatoid arthritis synovial fluid single strand RNA induces transcription of TNFα in monocytes. Ann. Rheum. Dis. 72(3), 418–426 (2013).
    • 37. Hu W, Yen Y-T, Singh S, Kao C-L, Wu-Hsieh BA. SARS-CoV regulates immune function-related gene expression in human monocytic cells. Viral Immunol. 25(4), 277–288 (2012).
    • 38. De Marcken M, Dhaliwal K, Danielsen AC, Gautron AS, Dominguez-Villar M. TLR7 and TLR8 activate distinct pathways in monocytes during RNA virus infection. Sci. Signal. 12(605), 1347(2019).
    • 39. Eriksson M, Meadows SK, Wira CR, Sentman CL. Endogenous transforming growth factor-β inhibits toll-like receptor mediated activation of human uterine natural killer cells. Am. J. Reprod. Immunol. 56(5–6), 321–328 (2006).
    • 40. Joetham A, Takada K, Taube C et al. Naturally occurring lung CD4+ CD25+ T cell regulation of airway allergic responses depends on IL-10 induction of TGF-β. J. Immunol. 178(3), 1433–1442 (2007).
    • 41. Ostroukhova M, Seguin-Devaux C, Oriss TB et al. Tolerance induced by inhaled antigen involves CD4+ T cells expressing membrane-bound TGF-β and FOXP3. J. Clin. Investig. 114(1), 28–38 (2004).
    • 42. Huang C, Wang Y, Li X et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395(10223), 497–506 (2020).
    • 43. Wan S, Yi Q, Fan S et al. Characteristics of lymphocyte subsets and cytokines in peripheral blood of 123 hospitalized patients with 2019 novel coronavirus pneumonia (NCP). Medrxiv (2020). • Explores the cellular immunity and cytokine status of patients with SARS-CoV-2 associated pneumonia.
    • 44. Zheng Y, Sun L, Jiang T, Zhang D, He D, Nie H. TNFα promotes Th17 cell differentiation through IL-6 and IL-1β produced by monocytes in rheumatoid arthritis. J. Immunol. Res. 2014, 2014 (2014).
    • 45. Hernandez-Pando R, Rook G. The role of TNF-alpha in T-cell-mediated inflammation depends on the Th1/Th2 cytokine balance. Immunology 82(4), 591 (1994).
    • 46. Tabarkiewicz J, Pogoda K, Karczmarczyk A, Pozarowski P, Giannopoulos K. The role of IL-17 and Th17 lymphocytes in autoimmune diseases. Arch. Immunol. Ther. Exp. 63(6), 435–449 (2015).
    • 47. Mcgeachy MJ. GM-CSF: the secret weapon in the T H 17 arsenal. Nat. Immunol. 12(6), 521 (2011).
    • 48. Smeltz RB, Chen J, Ehrhardt R, Shevach EM. Role of IFN-γ in Th1 differentiation: IFN-γ regulates IL-18Rα expression by preventing the negative effects of IL-4 and by inducing/maintaining IL-12 receptor β2 expression. J. Immunol. 168(12), 6165–6172 (2002).
    • 49. Grifka-Walk HM, Giles DA, Segal BM. IL-12-polarized Th1 cells produce GM-CSF and induce EAE independent of IL-23. Eur. J. Immunol. 45(10), 2780–2786 (2015).
    • 50. Wei L, Laurence A, Elias KM, O'Shea JJ. IL-21 is produced by Th17 cells and drives IL-17 production in a STAT3-dependent manner. J. Biol. Chem. 282(48), 34605–34610 (2007).
    • 51. Ivashkiv LB. Cytokine expression and cell activation in inflammatory arthritis. Adv. Immunol. 63, 337–376 (1996).
    • 52. Schoggins JW, Wilson SJ, Panis M et al. Correction: corrigendum: a diverse range of gene products are effectors of the type I interferon antiviral response. Nature 525(7567), 144–144 (2015).
    • 53. Ruiz-Ortega M, Bustos C, Hernández-Presa MA, Lorenzo O, Plaza JJ, Egido J. Angiotensin II participates in mononuclear cell recruitment in experimental immune complex nephritis through nuclear factor-κB activation and monocyte chemoattractant protein-1 synthesis. J. Immunol. 161(1), 430–439 (1998).
    • 54. Ortego M, Bustos C, Hernández-Presa MA et al. Atorvastatin reduces NF-κB activation and chemokine expression in vascular smooth muscle cells and mononuclear cells. Atherosclerosis 147(2), 253–261 (1999).
    • 55. Arthur JSC, Ley SC. Mitogen-activated protein kinases in innate immunity. Nat. Rev. Immunol. 13(9), 679–692 (2013).
    • 56. Lim YX, Ng YL, Tam JP, Liu DX. Human coronaviruses: a review of virus–host interactions. Diseases 4(3), 26 (2016).
    • 57. Keshet Y. The MAP kinase signaling cascades: a system of hundreds of components regulates a diverse array of physiological functions. In: MAP Kinase Signaling Protocols (Second Edition). Seger R (Ed.). Humana Press, NJ, USA, 3–38 (2010).
    • 58. Lippi G, Plebani M. Laboratory abnormalities in patients with COVID-2019 infection. Clin. Chem. Lab. Med. 58(7), 1131–1134 (2020).
    • 59. Wang J-T, Sheng W-H, Fang C-T et al. Clinical manifestations, laboratory findings, and treatment outcomes of SARS patients. Emerg. Infect. Dis. 10(5), 818 (2004).
    • 60. Ko J-H, Park GE, Lee JY et al. Predictive factors for pneumonia development and progression to respiratory failure in MERS-CoV infected patients. J. Infect. 73(5), 468–475 (2016).
    • 61. Hamimi A. MERS-CoV: middle East respiratory syndrome coronavirus: can radiology be of help? Initial single center experience. Egypt. J. Radiol. Nucl. Med. 47(1), 95–106 (2016).
    • 62. Jeon J-S, Rheem I, Kim JK. C-reactive protein and respiratory viral infection. Korean J. Clin. Lab. Sci. 49(1), 15–21 (2017).
    • 63. Sun J, Xiao Y, Zhang M, Ao T, Lang S, Wang J. Serum inflammatory markers in patients with adenovirus respiratory infection. Med. Sci. Monit. 24, 3848 (2018).
    • 64. Van Drongelen V, Holoshitz J. Human leukocyte antigen–disease associations in rheumatoid arthritis. Rheum. Dis. Clin. 43(3), 363–376 (2017).
    • 65. Hajeer AH, Balkhy H, Johani S, Yousef MZ, Arabi Y. Association of human leukocyte antigen class II alleles with severe Middle East respiratory syndrome-coronavirus infection. Ann. Thorac. Med. 11(3), 211 (2016).
    • 66. Liu J, Wu P, Gao F et al. Novel immunodominant peptide presentation strategy: a featured HLA-A* 2402-restricted cytotoxic T-lymphocyte epitope stabilized by intrachain hydrogen bonds from severe acute respiratory syndrome coronavirus nucleocapsid protein. J. Virol. 84(22), 11849–11857 (2010).
    • 67. Cai S, Sun W, Li M, Dong L. A complex COVID-19 case with rheumatoid arthritis treated with tocilizumab. Clin. Rheumatol. 39(9), 2797–2802 (2020).
    • 68. Song J, Kang S, Choi SW et al. Coronavirus disease 19 (COVID-19) complicated with pneumonia in a patient with rheumatoid arthritis receiving conventional disease-modifying antirheumatic drugs. Rheumatol. Int. 40(6), 991–995 (2020).
    • 69. Perrot L, Hemon M, Busnel J-M et al. First flare of ACPA-positive rheumatoid arthritis after SARS-CoV-2 infection. Lancet Rheumatol. 3(1), e6–e8 (2021).
    • 70. Derksen VF, Kissel T, Lamers-Karnebeek FB et al. Onset of rheumatoid arthritis after COVID-19: coincidence or connected? Ann. Rheum. Dis. 1–2 (2021).
    • 71. Duret P-M, Sebbag E, Mallick A, Gravier S, Spielmann L, Messer L. Recovery from COVID-19 in a patient with spondyloarthritis treated with TNF-alpha inhibitor etanercept. Ann. Rheum. Dis. 79(9), 1251–1252 (2020).
    • 72. Luo P, Liu Y, Qiu L, Liu X, Liu D, Li J. Tocilizumab treatment in COVID-19: a single center experience. J. Med. Virol. 92(7), 814–818 (2020).
    • 73. Quartuccio L, Valent F, Pasut E, Tascini C, De Vita S. Prevalence of COVID-19 among patients with chronic inflammatory rheumatic diseases treated with biologic agents or small molecules: a population-based study in the first two months of COVID-19 outbreak in Italy. Joint Bone Spine 87(5), 439–443 (2020).
    • 74. Alzghari SK, Acuña VS. Supportive treatment with tocilizumab for COVID-19: a systematic review. J. Clin. Virol. 127, 104380 (2020).
    • 75. Lafaie L, Célarier T, Goethals L et al. Recurrence or relapse of COVID-19 in older patients: a description of three cases. J. Am. Geriatr. Soc. 68(10), 2179–2183 (2020).
    • 76. Zhou L, Liu K, Liu H. Cause analysis and treatment strategies of “recurrence” with novel coronavirus pneumonia (COVID-19) patients after discharge from hospital. Zhonghua Jie He He Hu Xi Za Zhi 43, E028–E028 (2020).
    • 77. Dougados M. Comorbidities in rheumatoid arthritis. Curr. Opin. Rheumatol. 28(3), 282–288 (2016).
    • 78. Goudouris ES. Laboratory diagnosis of COVID-19. Jornal de pediatria 97(1), 7–12 (2021).
    • 79. Farng E, Friedrich JB. Laboratory diagnosis of rheumatoid arthritis. J. Hand Surg. (Am) 36(5), 926–927 (2011).
    • 80. Sinha N, Balayla G. Hydroxychloroquine and COVID-19. Postgrad. Med. J. 96(1139), 550–555 (2020).
    • 81. Funnell S, Dowling W, Muñoz-Fontela C et al. Emerging preclinical evidence does not support broad use of hydroxychloroquine in COVID-19 patients. Nat. Commun. 11(1), 1–4 (2020).
    • 82. Femere R, Aronson J. Chloroquine and hydroxychloroquine in COVID-19. 2020. BMJ 369, m1432 (2020).
    • 83. Sarzi-Puttini P, Giorgi V, Sirotti S et al. COVID-19, cytokines and immunosuppression: what can we learn from severe acute respiratory syndrome? Clin. Exp. Rheumatol. 38(2), 337–342 (2020). • Explores the possibilities of use of immunosuppressants in the new pandemic situation based on available information of prior pandemics (SARS-CoV and MERS-CoV).
    • 84. Singhal T. A review of coronavirus disease-2019 (COVID-19). Indian J. Pediatr. 87(4), 281–286 (2020).
    • 85. Qin C, Zhou L, Hu Z et al. Dysregulation of immune response in patients with COVID-19 in Wuhan, China. Clin. Infect. Dis. 71(15), 762–768 (2020).
    • 86. Wang F, Nie J, Wang H et al. Characteristics of peripheral lymphocyte subset alteration in COVID-19 pneumonia. J. Infect. Dis. 221(11), 1762–1769 (2020).