Skip to main content
Drug Design, Development and Therapy logoLink to Drug Design, Development and Therapy
. 2024 Sep 20;18:4215–4240. doi: 10.2147/DDDT.S475005

Cytokine Storm-Induced Thyroid Dysfunction in COVID-19: Insights into Pathogenesis and Therapeutic Approaches

Ali Attiq 1,, Sheryar Afzal 2,, Habibah A Wahab 1, Waqas Ahmad 1, Mahmoud Kandeel 2,3, Yassir A Almofti 2,4, Ahmed O Alameen 2,5, Yuan Seng Wu 6,7
PMCID: PMC11421457  PMID: 39319193

Abstract

Angiotensin-converting enzyme 2 receptors (ACE2R) are requisite to enter the host cells for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). ACE2R is constitutive and functions as a type I transmembrane metallo-carboxypeptidase in the renin-angiotensin system (RAS). On thyroid follicular cells, ACE2R allows SARS-CoV-2 to invade the thyroid gland, impose cytopathic effects and produce endocrine abnormalities, including stiff back, neck pain, muscle ache, lethargy, and enlarged, inflamed thyroid gland in COVID-19 patients. Further damage is perpetuated by the sudden bursts of pro-inflammatory cytokines, which is suggestive of a life-threatening syndrome known as a “cytokine storm”. IL-1β, IL-6, IFN-γ, and TNF-α are identified as the key orchestrators of the cytokine storm. These inflammatory mediators upregulate transcriptional turnover of nuclear factor-kappa B (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), and mitogen-activated protein kinase (MAPK), paving the pathway for cytokine storm-induced thyroid dysfunctions including euthyroid sick syndrome, autoimmune thyroid diseases, and thyrotoxicosis in COVID-19 patients. Targeted therapies with corticosteroids (dexamethasone), JAK inhibitor (baricitinib), nucleotide analogue (remdesivir) and N-acetyl-cysteine have demonstrated effectiveness in terms of attenuating the severity and frequency of cytokine storm-induced thyroid dysfunctions, morbidity and mortality in severe COVID-19 patients. Here, we review the pathogenesis of cytokine storms and the mechanisms and pathways that establish the connection between thyroid disorder and COVID-19. Moreover, cross-talk interactions of signalling pathways and therapeutic strategies to address COVID-19-associated thyroid diseases are also discussed herein.

Keywords: COVID-19, cytokine storm, signalling pathways, inflammation, thyroid disorders, cross talks

Introduction

In the initial stages of COVID-19, the patient exhibits anosmia, dyspnoea, non-productive cough, intermittent fever, and obstructive sleep apnoea.1 However, as the disease progresses, patients experience delirium, depression, ageusia, epigastric distress, thromboembolism, metabolic irregularities and acute respiratory distress syndrome (ARDS).2,3 Angiotensin-converting enzyme 2 (ACE2) receptors are requisite for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) to enter the host cells. It is worth mentioning that ACE2 receptors are constitutive and function as type I transmembrane metallo-carboxypeptidase in the renin-angiotensin system (RAS). Detail transcriptome analyses have identified ACE2 expressions on multiple organs, including pulmonary epithelium, kidney vasculature, myocardium and thyroid follicular cells, vindicating the plethora of pulmonary and extra-pulmonary manifestations in advanced COVID-19 patients.4

Studies suggest that ACE2 receptor interaction in COVID-19 involves structural binding with SARS-CoV-2 spike proteins, modulation by glycans, and post-infection regulation affecting immune response.5,6 ACE2 receptors on thyroid follicular cells allow SARS-CoV-2 to invade the thyroid gland, impose cytopathic effects and produce endocrine abnormalities. Clinical data suggest that COVID-19 patients experience various thyroid complications such as subacute thyroiditis,7,8 autoimmune thyroiditis,9,10 and abnormal thyroid function,11,12 with symptoms such as stiff back, neck pain, fever, muscle ache, lethargy, enlarged and inflamed thyroid gland.12–14 Further damage is perpetuated by the sudden burst of pro-inflammatory cytokines in the infected cell, which suggests a life-threatening syndrome known as a “cytokine storm”.15 Numerous immune effector proteins such as interleukins (ILs), interferons (IFNs), tumour necrosis factor (TNF), chemokines orchestrate the cytokine storm by upregulating the transcriptional activities of Janus kinase (JAK), signal transducer and activator of transcription (STAT), nuclear factor-kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK). Upon activation, these transcriptional factors momentously increase the plasma level of the inflammatory mediators, which hastens the disease progression by promoting systemic inflammation and multi-organ damage.16

SARS-CoV-2 infection induces thyroid follicular cell damage and hypothyroidism akin to its predecessor, SARS-CoV-1.11,17 According to recent studies, COVID-19 have the potential to actuate euthyroid sick syndrome (ESS), Hashimoto thyroiditis (HT), and Graves’ disease (GD) in patients with no previous history of thyroid disorders.18 Clausen, Rasmussen et al19 recently reviewed the similarities between inflammatory thyroid illness and COVID-19-induced hyper-inflammation. It is suggested that elevated IL-6 concentration perturbates the production and functionality of thyroid-stimulating hormone (TSH), which paves the pathogenesis of thyrotoxicosis in COVID-19.20

According to a recent epidemiological study by Ashrafi, Hatami, Bidhendi-Yarandi, Panahi,21 thyroid dysfunction is frequently observed in patients with COVID-19, characterised by a notable prevalence of non-thyroidal illness syndrome (NTIS) and thyrotoxicosis. It is worth mentioning that a significant prevalence of 26% and 10% of NTIS and thyrotoxicosis has been reported among COVID-19 patients, respectively.21 Nonetheless, the diagnostic challenges of COVID-19 patients with thyroid complications are multifaceted. Subacute thyroiditis and alterations in thyroid function can mimic or exacerbate COVID-19 symptoms, complicating the patient diagnosis and prognosis.22–24

Moreover, diagnosing COVID-19 patients with thyroid complications is challenging due to overlapping symptoms and laboratory findings. Pre-existing thyroid conditions can worsen COVID-19 outcomes, requiring careful management.23,25,26 Thyroid complications in COVID-19 patients have significant knowledge gaps, particularly in the variability of prevalence rates and the underlying mechanisms of thyroid disorders.21,27 There is also a need for greater awareness among the public and healthcare providers regarding the link between COVID-19 and thyroid issues and improved clinical management protocols for affected patients.21,28,29 Here, we review the pathogenesis of cytokine storms and the mechanisms and pathways that establish the connection between thyroid disorder and COVID-19. Moreover, cross-talk interactions of signalling pathways and therapeutic strategies to address COVID-19-associated thyroid diseases are also discussed herein.

SARS-COV-2 and Cytokine Storm

The SARS-COV-2 contains four (4) structural proteins, including spike (S), membrane (M), envelope (E) and nucleocapsid (N).30 The S protein facilitates the viral attachment to host receptors (Figure 1). The SARS-CoV-2 requires S protein proteolytic processing and degradation to initiate the endocytic pathway.31 The S protein has two functional units, namely S1 and S2. The S1 subunit binds to the host cell receptor, whereas the S2 subunit fuses the virus with the host cellular membrane.32 The M protein defines the virion’s shape and allows the virus to interact with the N protein. The N protein consists of the N-terminal domain (NTD) and C-terminal domain (CTD), both of which interact with the viral RNA genome and encapsulate the genome into virions during the replication phase by interacting with non-structural protein (nsp) 3.33,34 The E protein arranges the viral assembly, initiates the release and plays a definitive role in the virulency and COVID-19 progression. The SARS-CoV-2 employs a receptor cognition process similar to its predecessor, SARS-CoV. SARS-CoV and SARS-CoV-2 utilise ACE2 as a receptor to enter its target cells.35 However, Shang, Wan et al36 suggested that SARS-CoV-2 requires the transmembrane protease serine protease 2 (TMPRSS2), cathepsin L, and furin to gain entry into the host cells. It is noteworthy that ACE2 is co-expressed with TMPRSS2 in many tissues, including nasal epithelial cells, lungs, and bronchial branches, suggesting the tissue tropism of SARS-CoV-2.37

Figure 1.

Figure 1

SARS-CoV-2 virus structure.

The furin cleavage site formation in S protein subunits facilitates S-glycoprotein binding with the ACE2 receptor of SARS-CoV-2.38 The protease enzyme binds to the ACE2 receptor, cleaving the S protein into S1 and S2 subunits. This cleavage brings out irreversible conformational changes in the S protein, significantly increasing its affinity towards the ACE2 receptor.39 Zhou, Yang et al40 indicated that the rapid transmission, replication and infectivity of the SARS-CoV-2 results from the furin cleavage and the receptor-binding sites of the modified S1 subunit as the viral genomic messenger RNA (mRNA) enter the host cell cytoplasm together with the nucleocapsid. The E, M, and S proteins and several other accessory proteins are produced by translating this genomic mRNA.41 These proteins are inserted into the rough endoplasmic reticulum (RER) and transported to the endoplasmic reticulum-golgi intermediate compartment (ERGIC). The nucleocapsids prepared by N protein and genomic RNA fuse into ERGIC, allowing the infected cell to release the virus-containing vesicles through exocytosis or simply fusing with the plasma membrane.42 The receptor-binding domain (RBD) of the S1 subunit significantly contributes to the pathogenicity of COVID-19. The RBD S1 subunit on the S protein binds to the ACE2 receptors, which can be found abundantly in type-2 pneumocytes. Upon binding, the RBD S1 subunit downregulates ACE2 receptors and conversely upregulates the production of angiotensin-2 (AT2), which is well-known for its vasodilatory activity.43 As the AT2 production jacks up, it leads to tissue injury by increasing pulmonary vascular permeability.44 The SARS-CoV-2 comprises antigen-presenting cells (APC) that adhere to the host dendritic cells. Their interaction activates macrophages and other immune cells to bolster the production and release of pro-inflammatory cytokines. Excessive and uncontrolled production of cytokines and chemokines, including TNF-α, IL-1α, IL-6, IL-7, IL-8, IL-9, IL-10, G-CSF, GM-CSF, IFN- γ, IP-10, MCP-1, MIP-1β, PDGF and VEGF alter the cellular function, damages visceral tissues and create a perfect environment for systemic inflammation, which is suggestive of a life-threatening syndrome known as “cytokine storm”.45

Hypothalamic-Pituitary-Thyroid (HPT) Axis and COVID-19

While COVID-19 patients primarily exhibit pulmonary manifestations, the involvement and damage to other organs, including cardiovascular, lymphatic and endocrine systems, are translated as extrapulmonary manifestations.46 Of interest, the thyroid gland exhibits higher levels of ACE2 and TMPRSS2 expression than the pulmonary airways, which invariably allows the SARS-CoV-2 to quickly gain excess, target and pose direct cytopathic damage to the thyroid gland and the entire hypothalamic-pituitary-thyroid (HPT) axis. The said damage is often translated into thyroid dysregulations, including hypothyroidism, thyrotoxicosis, and non-thyroidal illness syndrome.3,15 Autoimmune thyroid disorders such as Graves’ disease have been reported in patients with no previous medical or family history of thyroid abnormalities.47 It is worth mentioning that spontaneous bursts of IL-1, IL-6 and TNF-α are mainly responsible for the development of thyrotoxicosis in critical care COVID-19 patients.48

SARS-CoV- and MERS-CoV-associated thyroid dysfunctions provide valuable insight into the SARS-CoV-2-induced HPT pathogenesis (Figure 2).49 Endocrine abnormalities are mainly due to aberrated pituitary-hypothalamus functions and insufficient thyroid-stimulating hormone (TSH) production and release. A substantial amount of neurons have also been found in the hypothalamus with cytoplasmic SARS genome sequences.50 Additionally, the number of TSH-positive cells reduces significantly in the pituitary region of SARS patients. The lower TSH levels are possibly linked to the alterations in TSH-secreting cells in the pituitary, leading to the interruption of the pituitary endocrine axis feedback loop.51 It is further suggested that COVID-19 patients experienced low TSH levels, and the degree of reduction is linked with COVID-19 disease severity.52 When comparing the serum TSH levels in COVID-19 and non-COVID-19 pneumonia patients with a similar degree of severity, the former showed a lower value of TSH, assuming that COVID-19 disease has a specific impact on the TSH-secreting cells. Few studies have suggested that it may have resulted from the direct cytopathic effect of SARS-CoV-2 on the pituitary gland and indirect damage by the induced cytokine storm.

Figure 2.

Figure 2

SARS-CoV-2-induced thyroid dysregulations. (a) The diagram illustrates the potential path of thyroid gland infection. ACE2, TMPRSS2, and αvβ3 integrin have been identified at all hypothalamic-pituitary-thyroid (HPT) axis levels. The SARS-CoV-2 enters thyrocytes and the HPT axis by binding to these receptors and facilitates direct viral attack to thyroid cells, thus causing thyroid dysfunctions. (b) The host lung-mediated infection triggers hyper-inflammatory immune responses that lead to the formation of reactive oxygen species (ROS) in epithelial cells. ROS promotes NLR pyrin domain containing 3 (NLRP3) and nuclear factor-kappa B (NF-κB) activation, which raises the number of cytokines and, thus, activates various signalling pathways in the cells. These immunological responses result in clinically significant conditions like cytokine storm and acute respiratory distress syndrome (ARDS). These affect thyroid cells and the HPT axis and induce thyroid diseases.

Cytokine Storm and Signal Transduction Pathways

Pattern-Recognition Receptor (PRR)

PRRs recognise the pathogens and damaged cells via interacting with pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), respectively.53 They appear phylogenetically before the emergence of adaptive immunity and thus are recognised as an impotent effector molecule of innate immunity. Depending on protein domain homology, the PRRs are classified into toll-like receptors (TLRs), retinoic acid-inducible gene-1 (RIG)-like receptors (RLRs), nucleotide oligomerisation domain (NOD)-like receptors (NLRs), C-type lectin receptors (CLRs) and absent in melanoma-2 (AIM2)-like receptors (ALRs).54,55

The TLRs and CLRs are extracellular PRRs that recognise pathogens in the endosomal compartment, while the rest serve as intracellular PRRs. Due to the presence of tandem caspase recruitment domain (CARD) in the N-terminal region, RIG-1 actively participates in the production of IFN following viral infections.56 The CARD reacts with the mitochondrial antiviral-signalling protein to recruit downstream signalling molecules such as TNF receptor-associated factor (TRAF) and inhibitors of NF-κB kinase (IκK), causing the IFN to be transcriptionally activated.57 Moreover, TLRs induce the signal transduction pathways by recognising the respective PAMPs and DAMPs, which results in the activation of NF-κB and MAPKs via the myeloid differentiation primary response 88 (MYD88) [29]. The findings are previously supported by Medzhitov, Preston-Hurlburt, Janeway,58 suggesting that the TLR4 activate NF-κB and CD80 expression, resulting in the release of numerous pro-inflammatory mediators such as cytokines, chemokines and reactive oxygen species (ROS).59

The thyrocyte functional PRRs, including TLRs and RLRs, are susceptible to PAMPs- or DAMPs- inducing cytokines and chemokines production.60 Harii, Lewis et al61 discovered that the PAMP induces adjuvant thyroid complications from detecting dsRNA by functional TLR3 in the human thyroid. The production of ILs, IFNs, and TNF inhibits thyroid function through MAPK and NF-κB activation. Furthermore, the innate immune response influences the thyrocytes to act as APCs, which recruit lymphocytes and provoke an autoimmune response, consequently developing hypothyroidism, ESS, GD and HT.62

Nuclear Factor-Kappa B (NF-κB)

NF-κB is an inducible transcription factor involved in various immuno-inflammatory processes. NF-κB signalling requires phosphorylation and degradation of inhibitory IκB kinase-β (IKKβ), which liberates the p50 and p65 dimers. These subunits are translocated into the nucleus, functioning as transcriptional factors and regulating the genes involved in the inflammatory processes.63,64 Giuliani, Bucci, Napolitano60 indicated that the PRR ligands, cytokines, T-cell and B-cell receptor signals, and MAPKs serve as NF-κB activating stimulus. Two signalling cascades activate NF-κB, namely canonical and non-canonical pathways. The canonical pathway is characterised by activating p50, p65, and c-Rel dimer transcription factors, encoding the expression of cytokines, chemokines, and adhesion molecules responsible for recruiting and activating inflammatory cells. The latter predominantly stimulates the p52/RelB transcription factor and upregulates the proteins required for B cell viability, function, and antigen presentation.45,65

Thyroid cells predominantly express TLRs and RLRs. Hence, it is unsurprising that NF-κB dysregulation plays a pivotal role in the pathogenesis of thyroid disorders. The constitutive activation of the NF-κB pathway results in aberrant cytokine gene expressions, especially IL-1β, IL-6, and TNF-α that act as the primary mediators of thyroid diseases, including ESS and autoimmune thyroid diseases (AITDs).66 Downstream NF-κB) signalling converts pro-caspase-1 to active caspase-1 that cleaves pro-IL-1β. Subsequently, the IL-1β generates ROS intracellularly and causes thyroid cell apoptosis and hypothyroidism. Kwakkel, Wiersinga, Boelen67 hypothesised that NF-κB-induced IL-1β leads to the degradation of TRβ1 mRNA and increases transcriptional repression of the TRβ1, causing D1 level to plunge in thyroid cells. Moreover, NF-κB-stimulated IL-6 production induces Th17 lineage transcription factor RAR-related orphan receptor-γ (RORγt) and differentiates Th0 lymphocytes into Th17 cells.68 The Th17 cells intensify intracellular IL-17 production, enhancing SARS-CoV-2 and thyrocyte survival by preventing cytotoxic T cells from destroying target cells and paving the way for AITDs.69 Regulatory T (Treg) cells are essential for immune responses to restrict chronic inflammation and autoimmunity.70,71 Upon T-cell receptor (TCR) stimulation, c-Rel induces FoxP3 (master transcription factor for Treg), which indicates that NF-κB canonical signalling plays a significant role in mediating Treg cell development and regulation.72

Janus Kinase (JAK)/Signal Transducer and Activator of Transcription (STAT)

JAKs are non-receptor-associated tyrosine kinases, that consist of JAK1, JAK2, JAK3 and tyrosine kinase 2 (TYK2). They are essential in transmitting extracellular signals from cytokine activating the STAT pathway.73 IL-6 and IL-23 stimulate STAT3 via JAK2, while IL-21 activates STAT1, STAT3 and STAT5 via JAK1 and JAK3.74 The members included are STAT1, STAT2, STAT3, STAT4, STAT5 and STAT6. Each protein has different functions in innate and adaptive immunity.75 STAT1 proteins are involved in producing vascular cell adhesion molecules (VCAM) and inducible nitric oxide synthase (iNOS).76 Additionally, STAT1 and STAT2 mediate IFN signalling; STAT1 is associated with type I and II IFN signalling, whereas STAT2 is the type I IFN mediator.77 STAT3 induces IL-6 and IL-10, whereas STAT4 and STAT6 are involved in adaptive immunity. STAT4 is utilised for T-helper (Th) 1 cell differentiation associated with autoimmune disorders. In contrast, STAT6 participates in Th2 cell differentiation, which causes an allergic reaction, namely airway hyper-responsiveness. Lastly, STAT5 regulates lymphoid proliferation, inhibiting apoptosis and inducing cytokines.71,75 Recent studies have suggested a strong correlation between the STAT pathway and COVID-19 pathogenesis. STAT4-mediated Th1 response, cytotoxic effect in specific plays a pivotal role in SARS-CoV-2 immunity in moderate to severe conditions. This is further supported by the reports suggesting the presence of virus-specific CD8+ T lymphocytes in approximately 70% of COVID-19 patients.78

The JAK/STAT is essential for mediating cytokine receptors’ signalling cascades. Cytokine binding at the extracellular domain initiates the cross-phosphorylation of JAK, activating tyrosine residues at the corresponding receptor. The activated JAKs create a docking site for STAT molecules, which paves the way for the dissociation, dimerisation, and nuclear translocation of phosphorylated STAT molecules.79 Among the family members, the function of STAT3 is underlined. IL-6-induced JAK/STAT3 activates the gp130/cytokine-specific receptor complex.80 Activated receptor complexes with STAT3 mediate IL-6 and IL-23 signals, which allows TH17 cells to differentiate and function as effectors. Likewise, the Th0 lymphocyte differentiates into Th17, upregulating the IL-17 expression, an essential mediator of autoimmune diseases, including GD.81 Kotkowska, Sewerynek, Domańska, Pastuszak-Lewandoska, Brzeziańska82 support the involvement of STAT3 polymorphism in the development and progression of AITD.

Moreover, combining IL-6 and JAK/STAT3 is a prerequisite for fully activating NF-κB. The nuclear translocation of phosphorylated STAT3 invariably promotes cytokine storm by NF-κB activation and abrogating IFN-γ production.66 NF-κB activation promotes IL-6 positive feedback loop mechanisms and IL-6 amplifier (IL-6 Amp) activation, which paves the way for major histocompatibility complex (MHC) class II-associated autoimmune disease, including GD.83 Lee, Nakagiri et al84 discussed that IL-6 Amp mainly targets C-C motif chemokine ligand 2 (CCL2), and the elevated concentration is apparent after IL-6 Amp activation. The increased CCL2 plasma level modulates macrophages’ cellular adhesion and chemotaxis through β1 integrins and p38MAPK signalling pathways.85

Mitogen-Activated Protein Kinases (MAPKs)

MAPKs are essential in transducing the signals from the surface receptors to the transcriptional factors. It is activated by cytokines such as IL-1, IL-6, and TNF-α. The MAPK comprises three main groups: c-jun N-terminal kinase (JNK), p38, and extracellular signal-regulated kinases (ERKs).86 p38 MAPK upregulates the expression of IL-1, IL-6, IFN-γ and TNF-α. In addition, the activated p38 MAPK in CD4+ T cells has an essential role in Th17 cell function by regulating the production of IL-17.87 The ERK MAPK governs the synthesis of IL-6, IL-12, IL-23 and TNF-α and induces thyroid cell damage by increasing pro-inflammatory cytokine production from macrophages.88 It is worth mentioning that JNK MAPK activation has been associated with decreased TSH and increased T4 serum levels. It has been suggested that the upregulation of the DIO3 gene can be induced by MAPK activation and the TGFβ pathway, leading to reduced serum T3 and T4 levels.89 Other than that, the production of ROS, specifically anion superoxide from TLR, increases the expression of intracellular adhesion molecule-1 (ICAM-1) by activating the MAPK pathway.90,91 The ICAM-1 is expressed by IFN-γ through JAK/STAT signalling, resulting in the transmigration of leukocytes to the inflamed site and co-stimulation for T cell activation. Based on these studies, it is highly suggested that MAPK signalling pathways are essential to stimulate thyrocyte destruction, which plays a significant role in the pathogenesis of AITD.92

Key Orchestrators of Cytokine Storm-Related Thyroid Complications in COVID-19

Interleukins (IL)

ILs are a cytokine group that regulates cell growth, differentiation, and immune activation during inflammation and other immune responses. These proteins induce various cells and tissues by binding to high-affinity cell surface receptors.65 The histo-biochemistry analysis of COVID-19 patients has revealed elevated levels of numerous cytokines (Table 1). In inflammation, IL-6 utilises cis- and trans-signalling to activate the JAK/STAT3 pathway.93 In cis-signalling, the IL-6 binds to gp130 and membrane-bound IL-6 receptor (mIL-6R) and activates downstream signalling of JAK/STAT3 and MAPK. This signalling cascade promotes the differentiation of Th17, CD8+ T and B cells and supports neutrophil migration. These actions further enhance IL-6 release and aggravate inflammation. In the trans-signalling, the circulating IL-6 binds to the soluble IL-6 receptor (sIL-6R) and induces JAK/STAT3 signalling in the cells lacking mIL-6R expressions, like endothelial cells and vascular smooth muscle cells (Table 1). The over-activation of JAK/STAT3 signalling triggers more mediators to be secreted, like IL-6 itself, thereby promoting C-reactive protein (CRP) in hepatocytes.94 Croce, Gangemi et al95 proposed that the elevated circulating IL-6 reduces Tg and T3 synthesis in thyroid follicular cells, leading to thyroid complications.

Table 1.

Mechanism of Action and Functions of Soluble Inflammatory Mediators in COVID-19-Related Thyroid Disorders

Mediators Mechanism of Action and Function (s)
Cytokines and growth factors
IL-1β Downregulates the expression of DIO1, decrease iodide and T3 uptake, induce Fas and MCP-1 mRNA levels in thyrocytes67,95
IL-2 Restricts EGF-stimulated thyroid follicular cell proliferation and induce thyroid autoantibodies production96
IL-4 Promotes Th2 cell differentiation and ameliorate Th1 response96
IL-5 Induce B cell proliferation97
IL-6 Downregulates TBG gene expression, decrease D1 and D2 activity by stimulating ROS, reduces T3 synthesis, rT3 clearance, and intracellular T4 to T3 conversion. Increase DIO3 gene and D3 activity and differentiate CD4+ T cells into Th17 cells74,98
IL-8 Increase the neutrophils adherence to endothelial cells99
IL-10 Promote autoreactive B cell survival through Bcl-2 stimulation100
IL-12 Prime towards IFN-γ stimulation and Th1 response101
IL-13 Induce B cells to produce IgG102
IL-15 Promote anti-apoptotic activity and lymphocyte survival103
IL-17 Stimulate the release of IL-1β, TNF-α and chemokines Promote anti-apoptotic activity by interfering with the Fas/FasL pathway in thyrocytes104
IL-23 Induce Th0 differentiation to Th17105
IFN-α and IFN-β Decrease iodine incorporation, TPO, and Tg gene expression in the thyrocytes106
IFN-γ Decrease Tg expression and production in thyroid cells Upregulate MHC class II on thyroid follicular cells and induce Fas expression on thyrocytes107
TGF-β Induces apoptosis and inhibit thyrocyte growth. Induce Th17 differentiation by stimulating IL-6 production108
TNF-α Decrease D1 activity, and T4 to T3 conversion. Decrease TSH-stimulated TPO and Tg gene expression in thyrocytes. Induce TRAIL-mediated cell apoptosis103
Chemokines
CXCL9 (MIG) Recruitment of macrophages, Th1 cells, NK cells, dendritic cells and strengthen CXCL10 activity104
CXCL10 (IP-10) Amplify Th1 immune responses and IFN-γ production109
CXCL11 Mimics the mechanism and actions of CXCL945
CCL2 (MCP-1) Th0 cells Polarisation to Th2 phenotype. Recruit monocytes and T cells at the inflamed thyroid gland108
CCL3 (MIP1α) Chemotactic agent; selectively attract CD8+ T cells108
CCL4 (MIP1β) Chemotactic agent; selectively attract CD4+ T cells108
CCL20 Activate IL-17 signalling and migrate Th17 cells to trigger a self-destructive immune response110

The IL-1 family upregulates T and B lymphocyte maturation, leukocyte adhesion and CRP production. The IL-1 affects nearly every tissue in the human body but selectively participates in thyroid-driven autoimmunity. IL-1 comprises two agonists: IL-1α and IL-1β. The latter is a well-studied member of this family owing to its pivotal role in autoimmunity.111,112 The Fas ligand is generally expressed on thyroid follicular cells, which allows them to invade and eliminate Fas-sensitive target cells. As a result, IL-1β exposure induces thyroid follicular cell apoptosis, implying that IL-1β-induced Fas expression is critical for thyrocyte destruction and hypothyroidism.113 Croce, Gangemi, Ancona, Liboà, Bendotti, Minelli, Chiovato95 also reviewed the role of IL-1β in the pathogenesis of thyroid dysfunctions, proposing that IL-1β reduces T3 secretion and Tg mRNA expression in the thyroid cells. Macrophages primarily secrete the IL-1β through apoptosis and pyroptosis. Aside from the functions mentioned earlier, IL-1β is one of the dominant activators of IL-6 expression and the NF-κB pathway to establish positive feedback for its production.94 Further, the inactivated IL-1β is cleaved by the NLR pyrin domain containing 3 (NLRP3) protein to initiate sustained inflammation, suggesting that the NLRP3-IL-1β signalling pathway is actively involved in the inception of cytokine storm in COVID-19 patients.93

Tumour Necrosis Factor (TNF)

TNF produced by macrophages plays a pivotal role in the pathogenesis of cytokine release syndrome. TNF is classified into TNF-α and -β based on functional variability (Table 1).114 TNF-α primarily binds to TNFR1 and TNFR2. According to Yang, Wang, Brand, Zheng,115 TNF-α and TNFR1 complex is infamous for modulating inflammatory pathways and promoting the adhesion and permeability of endothelial cells. TNF-α is a well-known inducer of NF-κB that initiates the immune cell hyper-activation in cytokine storm by inducing epithelial cell death and aggravating systemic inflammation.45 The late study of Lee, Lombardi et al116 on thyroid cell CD40+ signalling contributed to a better understanding of NF-κB role in thyroid disorders. CD40+, a TNF receptor expressed on the APCs and thyroid cells, is required for adaptive immune response.117 The overexpression of CD40+ on thyrocytes is associated with autoimmune disease. The activated CD40+ upregulates p65 and p52 subunits to initiate the NF-κB canonical and non-canonical pathways.60 The elevated plasma level of TNF-α decreases the DIO1 activity, T3 and Tg production, and TSH-stimulated TPO gene expression in the normal thyroid follicular cells, resulting in AITD.95

Chemokines

Chemokines are chemotactic cytokines essential for migrating and positioning immune cells in tissues. Chemokines can be divided into homeostatic and pro-inflammatory chemokines. The former controls the cell migration for normal tissue maintenance, while the latter directs the immune cells towards inflamed sites.118 Studies have suggested that IFN-γ-induced chemokines like CXCL9, CXCL10 and CXCL11 provoke various thyroid disorders (Table 1).109 Interestingly, all three chemokines bind to the same receptor, CXCR3. The CXCR3 is abundantly expressed by activated Th1 and CD8+ lymphocytes and is hypothesised to have an active role in the recruitment of immune cells to sites of inflammation. It has three isoforms, CXCR3-A, CXCR3-B, and CXCR3-alternative (CXCR3-alt). The activated CXCR-3A results in cell proliferation, migration and invasion via MAPK/ERK and MAPK/JNK signalling. Meanwhile, the activation of CXCR-3B triggers cAMP formation and apoptosis through the MAPK/p38 pathway.119 In addition, the attraction of high levels of CXCR3-expressing macrophages by CXCL10 promotes IL-6 production, which is responsible for the further deterioration of the thyroid cell.120

Interferons (INFs)

Infected cells produce IFNs as alarmins to stimulate intracellular defence against viral invasion. The IFN family can be categorised into IFN-α, IFN-β, and IFN-γ. IFN-α and IFN-β are considered type 1 IFN, whereas IFN-γ is called type 2 IFN. Their action on the neighbouring cells prevents virus replication and regulates immune response.121 The role of IFN-γ is underlined in the COVID-19-related cytokine storm, which activates protective immunity against the virus via JAK/STAT1 signalling and downstream the STAT1-IFN-γ-activated site cascades (Table 1).93 Of note, the IFNs are found to induce cytokine-related thyroid dysfunctions. For instance, the IFN-α restricts iodine incorporation and thyroid hormone release, as well as decreases thyroid peroxidase (TPO), sodium iodide symporter (NIS) and thyroglobulin (Tg) gene expression in the thyrocytes.106 In addition, IFN-γ upregulates ICAM-1 on the thyroid cells, establishing cell-cell interaction and boosting the inflammatory processes.

Signalling Cross-Talks and Thyroid Disorders

Euthyroid Sick Syndrome (ESS)

ESS is commonly known as non-thyroidal illness or low T3 syndrome. It is characterised by reduced triiodothyronine (T3) and thyroxine (T4) serum levels and elevated reverse T3 (rT3) with abnormally low serum TSH concentrations.122 According to de Vries, Fliers, Boelen,123 cytokines generated during viral infections can provoke ESS due to their potential to inflict numerous genes in thyroid hormone metabolism. Zou, Wu et al124 revealed that 41 out of 149 (28%) were diagnosed with ESS in moderate to severe COVID-19 patients. As a result, it is conceivable that COVID-19 has the potential to induce ESS. In this context, the cytokines secreted profoundly as a generalised reaction to the infection have drawn particular interest. Available findings have suggested that IL-1β, IL-6, and TNF-α are the vital inflammatory mediators that play pivotal roles in ESS pathogenesis.

SARS-CoV-2 invades epithelial tissue and stimulates local innate immune cells, inducing the production of inflammatory cytokines (Figure 3). Specifically, IL-6 produced by monocytes, macrophages, and dendritic cells serves as a key activator of the JAK/STAT3 by utilising the cis-signalling pathway.93 The IL-6 binds to mIL-6Rs expressed on immune cells and establishes an IL-6/IL-R/gp130 complex, initiating IL-6 signal transduction. Based on Wajner, Goemann, Bueno, Larsen, Maia88 study, all three deiodinase mRNAs involved in the p38 and ERK MAPK pathways are upregulated by IL-6. Upon the formation of the receptor complex, JAK/STAT initiation leads to MAPK cascade activation, stimulating the deiodinase gene transcription inducer called cAMP response element-binding protein (CREB).125 Kester, Kuiper, Versteeg, Visser126 investigation revealed that cAMP is one of the main factors that induce D3 activity. All three deiodinases are selenocysteines, and thiols contain oxidoreductases, making them equally susceptible to redox reactions.

Figure 3.

Figure 3

Proposed mechanism for the effects of cytokines on deiodinase function leading to euthyroid sick syndrome (ESS). (a) IL-6 binds to its receptor followed by the formation of the IL-6/mIL-6R/gp130 complex, initiates JAK/STAT3 pathway, p38 and ERK mitogen-activated protein kinase (MAPK) signalling. These MAPK cascades activate cAMP response element-binding protein (CREB) which induces DIO3 gene transcription. The increased D3 in thyrocytes further reduce plasma T3 and increase production of rT3 from T4. Additionally, TLR4/IL-1/MYD88-dependent pathway directs the downstream activation of transforming growth factor-β-activated kinase 1 (TAK1), stimulating IκB kinase (IKK) and MAPK signalling. This results in a synergistic increase in IL-1β by upregulating NLRP3 and pro-IL levels. The activation of NLRP3 cleaves pro-caspase-1 into active caspase-1 and causes pyroptosis, which leads to the secretion of IL-1β that can bind to IL-1R and stimulate MYD88 pathway. Hence, a vicious cycle of NLRP3 priming, activation, and pyroptosis that worsens ESS. Both excessive produced IL-1β and IL-6 also binds to NADPH oxidase 2 (NOX2) to generate ROS via the NADPH oxidase pathway. This process is catalysed by NADPH oxidase, which releases two electrons (e) to convert NADPH into NADP+. The molecular oxygen (O2) accepts the e and generates the ROS superoxide (O2-). ROS itself promotes NLRP3 inflammasome activation and depletes intracellular glutathione (GSH) thereby diminishing D1 catalytic activity. (b) Excessive TNF-α production activates NF-κB signalling and increases intracellular nitric oxide synthase (iNOS) activity in the liver, thereby producing large amount of ROS and hepatic oxidative stress. Since conversion of T4 into T3 primarily occurs in the liver, oxidative stress and diminished GSH levels impair D1 and D2 function, resulting in decreased T4 to T3 conversion and rT3 clearance. Taken together, excessive cytokine production particularly IL-1β, IL-6, and TNF-α activates multiple signalling pathways to disrupt deiodinase functions by reducing T3 levels while elevating rT3 concentrations observed in the ESS.

Interestingly, the elevated D3 function is directly associated with MAPK-induced DIO3 upregulation. On the contrary, DIO1 and DIO2 gene expressions remain unaffected due to the absence of catalytic position in D1 and D2. On the other hand, the presence of catalytic positing in D3 facilitates its easy access to extracellular glutathione (GSH) (Figure 3). In this scenario, if the intracellular concentration of GSH falls, D3 actions are not changed and remain unaffected.127 Conclusively, augmented D3 activity lowers plasma T3 levels by increasing T3 and T4 clearance and elevating rT3 conversion from T4.

From another perspective, the ESS is characterised by oxidative stress imposed by the production of ROS and reactive nitrogen species (RNS). The patients frequently exhibit lower antioxidant molecules (GSH) and lower reactions of the antioxidant enzymatic system associated with ROS detoxification.128 Wajner, Maia98 proposed that the elevated serum IL-6 level is directly linked to oxidative stress. The IL-6 triggers an oxidative burst by increasing superoxide radical production via nicotine adenine dinucleotide phosphate (NADPH) oxidase pathway activation.129,130 Consequently, intracellular GSH is depleted, and the GSH/GSSG ratio is lowered, affecting deiodinase functions when the intracellular redox state is perturbated (Figure 3).131

Deiodinases are oxidoreductases that facilitate iodine removal from the outer or inner ring of the thyroid hormones.132 Protein cysteine residues are oxidised to produce inter-and intramolecular disulfide bonds and GSH-mixed disulfides, especially important for deiodinase activity.133 Therefore, the raised ROS is expected to deplete putative thiol cofactors for D1 and D2, impairing catalytic reactions that demand a reductive intracellular environment. The IL-6-induced D1 depletion diminishes T3 synthesis and rT3 clearance. In contrast, the D2 shortage is complemented by disabling intracellular T4 to T3 conversion. Based on these reports, it is inferred that excessive IL-6 production results in decreased T3 plasma levels, which serve as a hallmark of ESS in COVID-19.

Furthermore, the ESS can be provoked through IL-1β and TNF-α activation during cytokine storm in COVID-19 patients.124 At the early stages of SARS-CoV-2 infection, the NLRP3-mediated inflammasome is activated by utilising two separate events. The first phase is priming, which involves the viral RNA engaging TLRs. The myddosome complex is recruited, which transmits downstream signals to NF-κB and elevates the levels of NLRP3 and pro-IL (Figure 3). The following step is assembling the inflammasome proteins into a functionally active structure comprising several signal molecules, creating an intracellular ion imbalance and promoting ROS generation.134 Typically, T3 regulates D1 mRNA expression by binding liganded thyroid hormone receptor (TR)-1 to the thyroid hormone response elements (TREs) in the DIO1 gene promoter region.

Meanwhile, the D1 mRNA activity is reduced due to the involvement of pro-inflammatory cytokines during illness. According to Kwakkel, Wiersinga, Boelen,67 a decline of D1 and TRβ1 mRNA levels by IL-1β is related to the NF-κB activation. It is hypothesised that the TRβ1 mRNA degradation is augmented during inflammation and transcriptional repression of the TRβ1 (through NF-κB) is due to the competitive behaviours of common factors in limited quantities to activate NF-κB and TRβ1-mediated D1gene transcription.

The Phagocytic NADPH oxidase pathway facilitates the IL-1β-induced ROS production, which resembles oxidative burst situ to IL-6-induced-ESS.135 TNF-α has been implicated in ESS due to its potent NF-κB activating potential. Nagaya, Fujieda et al136 discussed that the TNF-α-mediated-NF-κB activation disrupts the T3-dependent induction of deiodinase by reducing D1 promoter action, leading to the ESS. Further, it was suggested that thyroid hormone action is impaired in HepG2 cells since the liver is the primary site of T4 conversion to T3. These studies indicate that the cytokines, either alone or in combination, downregulate multiple TH production mechanisms, leading to lower T3 and T4 secretion from the thyroid gland.

Autoimmune Thyroid Disease (AITD)

AITD is known for its reactivity to self-thyroid antigens, resulting in destructive inflammatory reactions.109 Few documented incidences have reported severe hypothyroidism or GD during and after COVID-19 disease.47,137 Depending on the primary effector mechanism and type of cell implicated, it is classified as a T-cell or autoantibody-mediated disease. The T-cell infiltrates and destroys the target tissue, known as T-cell-mediated disorder, as seen in HT. In contrast, autoantibody-mediated disease is described as the disruption of function, as in GD.138 The cytokine productions are stimulated by thyroid follicular cells and are initially induced by IL-1, IFN-γ and TNF-α. They upregulate the expression of MHC class I and II on the surface of thyroid follicular cells to recruit T cells. Therefore, it is indicated that COVID-19-induced hyper-inflammation predisposes individuals to autoimmune complications (Figure 4).

Figure 4.

Figure 4

Proposed mechanism for COVID-19-associated cytokine storm-induce autoimmune thyroid diseases (AITDs), (a) The IL-4/STAT6/GATA3 axis regulates the differentiation of naïve CD4+ T-cells to Th2 cells. Excessive IFN-γ also stimulates Th2 cell differentiation through JAK/STAT1 pathway to transactivate class II trans activator (CIITA). MHC class II genes are transactivated by recruiting CIITA and CREB to their promoters. The MHC class II loaded within the peptide is then expressed on the cell surface to T-cell. The MHC class II engages with T-cell receptor (TCR) and activates PI3K/mTORC2/Akt pathways. The engagement promotes STAT6-induced GATA3 transcription, thus producing Th2 effector cells. The Th2 cells express CD40L bind to CD40+ on B cells to promote mass production of TSH receptor autoantibodies (TRAb). TRAb binds to epitopes in extracellular domain of TSHR, activates Gαs/AC/PKA/CREB, Gαq/PLC/PI3K/Akt, and Gαq/DAG/NF-κB pathways, which are pivotal for thyrocyte proliferation, function and survival. Upon SARS-CoV-2 binding to TLR4, MYD88 recruits and phosphorylates IRAKs. The phosphorylated IRAK then interacts with tumour necrosis factor receptor-associated factor 6 (TRAF6) to activate TAK1 and TAB. At the same time, TIR-domain-containing adaptor-inducing interferon-β (TRIF) serves as the adaptor of TLR3 and part of TLR4. TRIF interacts with receptor-interacting protein 1 (RIP1), causing its polyubiquination with TAB. Subsequently, IκB and MAPK are activated and translocate NF-κB and AP1 to the nucleus. The translocation upregulates inflammatory cytokines that worsens AITDs. Moreover, IL-6, IL-23, and TGF-β are the inducer of Th17 cell differentiation by stimulating STAT3 signalling pathways and RAR-related orphan receptor-γt (RORγt) transcription. IL-17 produced by Th17 cells binds to its receptor and recruits NF-κB activator 1 (ACT1) to initiate NF-κB signalling pathway. Activation of NF-κB by IL-17 results in enhanced Bcl-2 expression for thyrocyte survival. Thus, various signalling pathways synergistically regulate thyrocytes proliferation and survival by stimulating TRAb and cytokine productions, along with anti-apoptotic gene expression, contribute to the progression of Graves’ disease (GD). (b) Both IL-12 and IFN-γ activate STAT1 and STAT4 to stabilize T-bet expression. These pathways differentiate Th0 cells into Th1 cells, and produce Th1-specific cytokines like IL-1β, IFNs, and TNF-α. IL-1β itself promotes ROS production through NADPH oxidase pathway to damage thyroid cells. The combination of IL-1β and TNF-α sensitize thyrocytes to TRAIL-induced apoptosis by recruiting Fas-associated death domain (FADD) and MAPK signalling. The MAPK-activated caspase 8 and FADD-activated caspase 9 cleaves procaspase 3 to its active form by direct proteolytic processing, and a series of downstream events that lead to thyrocyte apoptosis. Meanwhile, combination of IL-1β and IFN-γ sensitize thyrocytes to Fas-induced apoptosis. IFN-γ binds to type II IFN receptor and stimulates JAK/STAT1 pathway. The activated STAT1 induces caspase 8 expression and thus initiating thyroid cell death. IFN-α binds to type I IFN receptor and triggers a signaling cascade predominantly driven by JAK/STAT. IFN-α increases oligoadenylate synthetase 1 (OAS1) expression which is an innate sensor of viral infection, CXCL10 for mononuclear cell recruitment, Tg and TPO surface expression. The excessive surface Tg and TPO act as autoantigens, and bind to CD16 of NK cells to enhance antibody-dependent cytotoxicity (ADCC). Perforin and granzymes are then released from NK cells into thyroid cells to cause cell injury. IFN-α also increases MHC class I expression presented on the cell surface to CD8+ cytotoxic T-cell. Combining the effects of cytokines, perforin, granzymes, and cytotoxic T cells on the thyroid cells, these propose COVID-19-related cytokine storm causes thyrocytes apoptosis and hypothyroidism, leading to Hashimoto thyroiditis (HT).

AITD patients have more CD4+ T cells, exhibiting abnormal FoxP3 expression, a critical gene for developing Treg cells. The Treg suppresses lymphocyte differentiation into Th1 cells.107 The reduced Treg activity enhanced follicular T helper cell function, and the DNA fragment release contributes to the initiation and perpetuation of HT.139 The HT is characterised by a Th1 immune response that favours cell-mediated immunity and apoptosis in thyrocytes. The Th1 cytokines are synthesised from the naïve Th cells by TCR engagement. In addition, IFN-γ binds to its cognate receptor to initiate STAT1 signalling. This phosphorylated STAT1 stimulates the expression of the transcription factor T-bet, which leads to the differentiation of Th cells into Th1 cells by trans-activating IFN-γ and receptor for IL-12. Subsequently, the cell becomes responsive to IL-12, and the signalling through STAT4 further stabilises the Th1 phenotype. The Th1 cells secrete pro-inflammatory cytokines such as IFN-γ, TNF-α, and IL-1β, which activate the macrophages and cytotoxic effects.

The thyroid peroxidase (TPO) autoantibody levels represent MHC expression on thyrocytes and the degree of lymphocyte infiltration, which stimulates autoantibody production.140 In patients with HT, anti-TPO antibodies depend on GSH levels, exhibiting an inverse correlation.141 The IFN-α restricts iodine incorporation and decreases the thyrocytes’ TPO and thyroglobulin (Tg) gene expression.106 IFN-α receptor binding and JAK/STAT activation stimulate the interferon-stimulated genes (ISGs). As such, the TPO surface expression on thyroid cells is dramatically increased by IFN-α. Subsequently, hyper-expression of TPO as thyroid autoantigens leads to TPO autoantibodies formation, mainly when IFN-α causes a reduction in T suppressor cells and elevation of B cells. As expected, IFN-α boosts the MHC class I antigen expression on the thyrocytes, thus recruiting cytotoxic T lymphocytes and causing inflammatory responses.142 Down the lane, TPO antibodies mediate thyroid cell damage by antibody-dependent cytotoxicity (ADCC). Autoantibodies attached to the thyrocytes recognise the Fc receptor on NK cells, which results in thyroid cell lysis.143 Among the mechanisms of ADCC involving NK cells, the perforin/granzyme pathway has been widely studied. Self-reactive CD4+ T cells attract CD8+ cytotoxic T cells and stimulate the release of cytotoxic granules containing perforin and granzyme to induce cell necrosis.144

TNF, FasL, and TRAIL pro-apoptotic death ligands and receptors are significantly expressed in thyrocytes, which are normally inactive in healthy settings.145 Interestingly, IL-1β, IFN-γ, and TNF-α activate the apoptotic pathways, typically inactive in thyroid cells. Mezosi, Wang et al103 suggested that combining IL-1β and TNF-α promotes TRAIL-mediated cell death by increasing TRAIL receptors and procaspase expression and decreasing p44/p42 MAPK pathway activity. This pathway has been reported to have a dominant protecting role against apoptotic signalling from death receptors. At the same time, the activation kinetics were slowed in the IL-1β/TNF-α-treated cells.146,147 Hence, declining p44/p42 MAPK signalling facilitates thyroid cell apoptosis. Mezosi, Wang et al148 again suggested that the signalling pathways that include everything from the receptor to the apoptosis-inducing caspase enzymes undergo profound alterations when the thyrocytes are exposed to the IL-1β together with IFN-γ. Although the IFN-γ upregulates the Fas receptor expression, contrarily, it does not sensitise the thyrocytes but abrogates Bid concentration, which is crucial for cell apoptosis and hypothyroidism. The GD promotes Th2, and naïve T cells are differentiated via STAT6 activation. The STAT6 is phosphorylated when TCR engages with the IL-4 receptor, leading to the Th2 transcription factor GATA3 induction. Th2-specific cytokines, including IL-4, IL-5, IL-6 and IL-13 are trans-activated by GATA3.149 At the same time, STAT4 and IL-12 receptors required by Th1 are down-regulated due to the overexpression of IL-6 (Figure 4).

The predomination of Th2 cytokines stimulates humoral immunity rather than cellular immunity, boosting the synthesis of B lymphocyte autoantibodies. The elevated immunoglobulin G (IgG) levels caused by the Th2 cytokines potentially downregulate Fas expression while upregulating anti-apoptotic molecules like Bcl-2.103 As a result, the overexpressed Bcl-2 in the thyroid cells induces GD, making them resistant to Fas/FasL-mediated apoptosis. On the other hand, this resistance is unlikely to be observed in thyroid-infiltrating lymphocytes, where the pro-apoptotic proteins are expressed.150 Thus, it indicates that the regulation of the thyroid microenvironment in GD is almost opposite to that of HT, where the infiltrating lymphocytes in GD are sensitive to apoptosis. Still, the thyroid cells escape cell death due to overexpression of anti-apoptotic molecules.

During SARS-CoV-2 infection, the NF-κB is activated by TLR on macrophages, dendritic cells and other innate immune cells, which operate as the first-line defence combined with anatomical barriers. The TLRs develop homodimers and recruit adaptor molecule MyD88, followed by the recruitment of IRAK to the receptor complex by MyD88’s death domain.151 Auto-phosphorylation of IRAK dissociates the complex and paves the way for TRAF6 recruitment. On top of that, the transforming-growth-factor-β-activated kinase 1 (TAK1) linked with adaptor TAB molecules stimulation inactivates IKK and phosphorylate IκB and initiates NF-κB nuclear translocation.152 This canonical pathway leads to the augmented production of cytokines such as IL-6, IL-8, and TNF-α, directly related to AITD development. As previously mentioned, viral infections trigger an autoimmune response linked to MHC antigen expression in the non-immune cell. According to Hanafusa, Chiovato et al,153 the IFN-γ produced in the target organ promotes human leukocyte antigen (HLA) class II expression in the epithelial cells for the first time. Subsequently, this allows the autoantigens to be presented and auto-reactive T cells to be activated. They revealed that such infection would aggravate first-time-MHC class II molecule expression on the thyroid follicular cells, and these cells could behave as APC to play a role in AITD induction. The increased T cells and cytokines would then perpetuate such thyroid disorders.

Upon recognising the presented peptide by the T cell receptor on the thyroid cell (particularly CD40+), send the co-stimulatory signals required for T cell activation [84]. CD40+ is a TNF receptor (TNF-R), a major immune-modulating susceptibility gene for GD. It is known to be expressed on APCs and thyroid cells and is essential for adaptive immunity.154 Furthermore, it interacts with CD154+ expressed on the T cells to give a co-stimulatory signal that induces autoreactive B cell proliferation, immunoglobulin class switching, and germinal centre development. Of note, the activation of CD40+ in B cells suppresses Fas from inducing apoptosis. Zazzeroni, Papa et al155 suggested that CD40+-mediated cytoprotection implies the activation of protective gene gadd45β via NF-κB/Rel transcription factors. Additionally, CD40+ in mature B lymphocytes stimulates pro-survival signalling by antagonising IgM-induced apoptosis in the immature B cells. As pro-inflammatory situations overtly drive B cell proliferation, pro-apoptotic cytokines activate the constitutive NF-κB signalling supporting thyrocytes proliferation and survival.

The overexpressed IL-6 during a cytokine storm induces a similar anti-apoptotic activity as CD40+, signalling in COVID-19. Th17 cells, a CD4+ T cell subset distinct from Th1 and Th2 cells, primarily generate IL-17, a potent inflammatory cytokine.156 The STAT3 mediates IL-6 signals to the Th17 cells, allowing them to differentiate and function as effectors. Subsequently, the Th0 lymphocyte differentiates into Th17, intensifying IL-17 synthesis in the cellular environment, an essential factor of autoimmune diseases.81 The synergistic interplay of IL-6 and IL-17 has been linked to viral persistence and worsening clinical outcomes during viral infection. Hou, Kang, Kim69 proposed that the additive effect of these interleukins induces the anti-apoptotic molecule Bcl-2, which prevents SARS-CoV-2-infected cells from being destroyed by virus-specific CD8+ T cells, allowing the virus to survive. Besides, it is indicated that IL-17 protects thyrocytes apoptosis by interfering with the Fas/FasL pathway, resulting in GD. Furthermore, the differentiation of Th17 into cells that perform various functions after prolonged exposure to various cytokines may indicate their substantial plasticity. For instance, after being exposed to IL-23 constantly, they develop into specialised pathogenic cells known as non-classic Th1, capable of producing IFN-γ.157 This, without a doubt, significantly influences the pathophysiology of molecular damage during AITD (Figure 4).

Lui, Lee et al158 suggested that three COVID-19 patients admitted to a non-intensive care unit presented detectable levels of TSH receptor autoantibodies (TRAb), at the same time, reduced serum TSH levels with normal fT3 and fT4, implying GD. The autoantibodies are produced due to loss of tolerance to TSHR, thus mimicking the action of TSH and causing hyperthyroidism [107]. This can be explained by the overexpression of the CD40+ signalling, which is demonstrated to increase TSHR antibodies. It is pivotal in adaptive immunity, especially in differentiating B cells into antibody-producing plasma cells.159 These autoantibodies, in turn, promote the proliferation and survival of thyroid follicular cells via the phosphatidylinositol 3,4,5 triphosphate kinase/protein kinase B (PI3K/Akt) pathway.160 It is worth mentioning that TSHR is a G-protein coupled receptor (GPCR) which stimulates the cAMP synthesis and PI3K/Akt signalling cascade after autoantibody binding. Although TSH and stimulatory autoantibodies trigger the same TSHR signalling pathway, the TRAb remains activated due to distinct pharmacodynamics and higher stability.161

Consequently, protein kinase A and CREB are activated, leading to inflammatory mediator synthesis and thyroid cell growth. Simultaneously, PIP3 is induced when G protein downstream signalling triggers the canonical pathway that phosphorylates Akt by 3-phosphoinositide-dependent protein kinase 1 (PDK1) to increase cell survival.162 Taken together, the TRAb-TSHR signalling pathway, which activates G proteins and various cascades, is critical for thyrocyte proliferation and progression of GD.

Thyrotoxicosis

In a recent report, Muller, Cannavaro et al163 claimed that 13 out of 85 (13.5%) patients exhibit varying degrees of thyrotoxicosis symptoms in COVID-19. It is characterised by decreased TSH and elevated T3 and T4 serum levels linked to excessive IL-6 production during COVID-19. The connection between thyrotoxicosis and the raised IL-6 levels in the individuals implies that the inflamed thyroid gland is a consequence of a COVID-19-associated cytokine storm, resembling thyroid diseases that arise during immunotherapy treatment.164 The increased concentration of IL-6 disrupts deiodinases and thyroid hormone transport proteins and impairs pituitary cell TSH secretion, leading to abnormal thyroid functional parameters.48 The THYRCOV study further supports this notion by concluding that patients with thyrotoxicosis exhibit abnormally higher levels of IL-6.20

Subacute thyroiditis (SAT) is a self-limiting thyroid disorder that results from a viral or post-viral inflammatory process.15 It is characterised by neck discomfort, which is the hallmark of the clinical syndrome, plus a triphasic phase of thyrotoxicosis, hypothyroidism, and restoration to normal thyroid function.165,166 Few studies have reported the incidence of SAT during or after the SARS-CoV-2 infection, thus raising the concern that physicians should consider COVID-19 an underlying cause in patients with SAT.22,167 As mentioned, the thyroid gland and the whole HPT axis might be emerging and critical targets of SARS-CoV-2 damage. The prevalence of ACE2 and TMPRSS2 receptors in the thyroid explains the pathology of COVID-19-induced SAT. According to Caron,48 thyrotoxicosis may be caused by thyrocyte lysis, which produces thyroid hormones, combined with a decline in deiodinase activity in COVID-19. The inflammatory deterioration of the thyroid gland renders colloid leakage from the injured follicles into the interstitial tissue and then into the circulatory system in the early stage. This follicular cell destruction is thought to release lysozymes, which decompose the colloid and release a range of iodinated compounds such as iodoproteins, proteases, and amino acids.168 As a result, this leads to a rise in T3 and T4 levels, which simulates hyperthyroidism in this phase.

Moreover, the patients with SAT present decreased levels of thyroxine-binding globulin (TBG), a critical thyroid hormone transport protein that binds and transports thyroid hormones to the respective tissues. It is well-known for keeping serum thyroid hormone at a constant level. When TBG levels drop, fT4 levels rise, and TSH is adversely inhibited. Therefore, TBG is responsible for preventing thyroid hormone fluctuations.169 IL-6 has a significant inverse relationship with TBG levels, suggesting that the rise in serum IL-6 decreases serum TBG levels. However, it is yet unclear how IL-6 affects TBG gene expression.

Therapeutic Opportunities and Challenges

With the rise of IL-6, the interaction of gp130 with cytokine-specific α-chain activates the immune cells and JAK/STAT and MAPK signalling cassettes, which subsequently actuates pro-inflammatory and ROS responses.125 ROS formation plays a significant role in deiodinase malfunction; hence, inhibiting oxidative stress could be a probable strategy to restore thyroid function. An endogenous GSH deficit has been hypothesised to be a critical contributor to this redox homeostasis imbalance, leading to severe manifestations in COVID-19 patients. Antioxidant and anti-inflammatory activities of N-acetyl-cysteine (NAC) have been utilised in clinical trials to treat individuals with COVID-19-associated thyroid dysregulations in conjunction with standard therapies.170 Wajner, Goemann, Bueno, Larsen, Maia88 investigation on the impact of IL-6 on endogenous cofactor-mediated deiodinase function showed that T3 production by D1 and D2 is reduced by IL-6, despite the rise in deiodinase expression. In their study, the administration of NAC prevented IL-6’s inhibitory activity by restoring intracellular GSH concentration, highlighting that pro-oxidants are involved in IL-6-mediated effects. To regenerate active enzymes, all three deiodinases utilise a reducing agent, a thiol or thiol-dependent molecule, to release iodine from the selenocysteine residue.171 The NAC reinstated deiodinase function by re-establishing intracellular cysteine levels and the enzyme thiol cofactor, restoring redox balance. Since D3 has been linked to ESS in clinical studies, the role of NAC in correcting ROS-induced D3 overexpression cannot be neglected.172

Total antioxidant capacity (TAC) is the ability to neutralise ROS, depending upon the extent of exogenous and endogenous antioxidants in the body fluids.173 A late study showed that NAC-treated patients exhibited lower carbonyl concentrations with improved TAC parameters.174 Moreover, NAC significantly suppresses NF-κB-driven oxidative stress and downregulates pro-inflammatory gene expressions.175 During infections, cytokine production can be attenuated by inhibiting ROS-dependent activation of NF-κB by TLR3/hemagglutinin. The NAC can protect proteins from oxidative damage while restoring the antioxidant potential. Bhattacharya, Mondal et al176 illustrated that moderate-to-severe COVID patients who administered NAC 1g IV with standard treatment exhibited better chances of survival, improved discharge rate and reduced need to transfer patients to the intensive critical unit.

The contribution of TNF-α to hyperinflammation of COVID-19 has been subject to study. While anti-TNF agents have yet to receive approval for COVID-19 treatment, research suggests their potential efficacy in severe cases.177–179 Notably, the Inflammatory Bowel Disease Registry (SECURE-IBD) has reported the advantageous effects of TNF-α blockers in treating COVID-19-affected patients with inflammatory bowel disease (IBD). Analysis of 2307 IBD patients undergoing anti-TNF-α monotherapy before and during COVID-19, alongside 2088 patients receiving sulfasalazine/mesalazine treatment, revealed that 8% in the anti-TNF-α group were hospitalised and <1% died, as opposed to 20% hospitalised and 3% deceased in the other group.178,180 Furthermore, the Global Rheumatology Alliance (GRA) registry affirmed the favourable impact of TNF-α blockers on COVID-19 treatment in rheumatoid arthritis (RA) patients. Among 1388 RA patients administered TNF-α inhibitors, 103 (7.4%) necessitated ventilation, with 36 (2.6%) fatalities. Conversely, other regimens such as rituximab and JAK inhibitors posed heightened risks of severe illness compared to TNF-α inhibitors. No significant associations were identified with abatacept or interleukin 6 inhibitors.181 Data suggests that TNF-α blockers are associated with reduced rates of COVID-19-related hospitalisation.182,183 Nevertheless, further investigations are warranted to elucidate the optimal timing of intervention and the potential implications on vaccine efficacy in severe COVID-19 cases.

Inactivation of NF-κB is a druggable targeted strategy to combat COVID-19-associated thyroid diseases. A synthetic glucocorticoid, dexamethasone, is an effective treatment in blocking the release of pro-inflammatory cytokines.184 According to Bessler, Mendel et al,185 the expression of IFN-γ, IL-1β, TGF-β and TNF-α are suppressed by dexamethasone, thus reducing cytokine-related thyroid complications. Additionally, as an immune modulator, it lowers the IL-6 synthesis and reduces the cytokine feedback of NF-κB.66 The dexamethasone upregulates inhibitory IκBα which restricts NF-κB nuclear translocation.186 On top of that, dexamethasone prevents NF-κB from activating by stoichiometrically binding to the glucocorticoid receptor (GR) in the nucleus.187 The GR suppresses the transactivation of targeted genes by binding to transcription factors, NF-κB p65, or interacting with alternative protein IKK. Since the phosphorylation of IKKβ is essential for NF-κB activation, preventing such a process can provide therapeutic benefits against systemic inflammation.188

The dexamethasone at 6 mg/day for ten days in the Randomised Evaluation of COVID-19 Therapy study (RECOVERY) showed decreased mortality in one-third of ventilated patients. However, it failed to show significant benefits in mild-to-moderate COVID-19 and in patients who do not require oxygen support.189 This strengthens the hypothesis that dexamethasone’s positive effects on severe COVID-19 patients may be partially attributed to its ability to prevent NF-κB activation. Moreover, dexamethasone directly influences the HPT axis by affecting TRH in the hypothalamus.190 A study by Alkemade, Unmehopa, Wiersinga, Swaab, Fliers191 discussed the usage of glucocorticoids to reduce TRH mRNA levels in the hypothalamus, which is the leading cause of decreased pituitary TSH secretion. Formerly, human serum TSH levels were documented to be influenced by glucocorticoids. Wilber, Utiger192 demonstrated that glucocorticoids in higher dosages lower TSH levels in both hypothyroid and healthy individuals. On the contrary,193 suggested that glucocorticoid reduces TSH release from thyrotropes in a protein kinase C-dependent way via the protein annexin 1 and does not cause central hypothyroidism that requires thyroid hormone replacement.

Furthermore, the TRH suppression in the hypothalamus shows how glucocorticoids affect TSH secretion. The presence of GR in the TRH neurons of the hypothalamic paraventricular nucleus (PVN) and the glucocorticoid response element in the TRH gene promoter.190 The ERK1/2 signalling is crucial for stimulating TRH mRNA levels in the hypothalamus.194 According to Zhao, Su et al,195 MAPK phosphatase-1 (MKP-1) negatively regulates MAPK through phosphate removal from tyrosine and threonine in ERK. Some researchers revealed that dexamethasone could stimulate its anti-inflammatory effect by increasing MKP-1 expression and reducing proteasomal degradation of MKP-1.196 Subsequently, the CREB phosphorylation, an essential pathway for TRH gene transcription, is suppressed by administering dexamethasone.197 Therefore, it is indicated that dexamethasone inhibits the TRH-TSH-T4 axis and simultaneously exhibits an anti-inflammatory effect in patients with COVID-19-related thyroid dysregulations.

As IL-6 is the significant JAK/STAT activator, targeting it seems to be a plausible strategy for treating COVID-19-associated thyroid disorders.198 The STAT3-mediated IL-6 and IL-23 signalling are critical for maintaining Th17 cell differentiation and activating STAT3 via JAK2; hence, it is postulated that employing JAK2 inhibitors can suppress the pro-inflammatory function of Th17 cells.75 Baricitinib, an ATP competitive kinase inhibitor, targets JAK1 and JAK2 by inhibiting the kinase activity and precluding STAT3 activation.199 Moreover, inflammatory signalling of IL-2, IL-12, IL-23, and IFN-γ are also reported to be obturated by baricitinib.200 Apart from reducing the inflammatory response, baricitinib also disrupts viral host cell entry and assembly by acting on ACE2 receptor regulators, including AP2-associated protein kinase-1 (AAK1) and cyclin G-associated kinase (GAK), which govern clathrin-dependent endocytosis.201

However, the idea of treating COVID-19-associated cytokine storm with baricitinib seems to be controversial and must be addressed accordingly. It is established that when a pathogen like SARS-CoV-2 invades, an inflammatory response will be incepted by the host cells as a defensive action. Given that the JAK/STAT signalling is responsible for incepting antiviral response (IFNs), employing JAK/STAT inhibitors seems illogical. According to recent reports, over 80% of COVID-19 asymptomatic cases do not require hospital admission and successfully clear the viral titter due to IFNs-driven endogenous antiviral potential; hence, it is not advised to administer baricitinib202 in this population. In patients with cytokine release syndrome and ascending viral load, JAK/STAT pathway inhibition could potentially address systemic inflammation and disrupt viral entry and assembly intracellularly. It is worth mentioning that baricitinib exhibits low plasma protein binding and CYP450 interaction, making it a suitable candidate for combination therapy for critical care patients.203 Remdesivir was previously repurposed during the outbreak to manage the Ebola virus, and yet again, it has been recently approved by the FDA for the management of COVID-19.204,205 It is a monophosphoramidate prodrug that targets viral RNA and functions as a broad-spectrum antiviral. Remarkably, owing to a CoV-specific proofreading exoribonuclease (ExON) expressed in nsp14; various antivirals appear ineffective against SARS-CoV-2.206 The remdesivir’s unique ability to bypass viral proofreading, integration into viral RNA, and the subsequent suppression of RNA-dependent RNA polymerases (RdRps) allows it to limit viral replication.207 Moreover, its maximum serum concentration (Cmax) can suppress 90% of SARS-CoV-2 replication, a significant parameter for effectively managing COVID-19.208

As with sepsis, the cytokine storm associated with COVID-19 is triggered by an infection. The efficacy of therapies targeting a single cytokine is limiting, as multiple cytokine signalling pathways are assumed to be involved in this process. Thus, combination therapies may be a better strategy for the following reasons. 1) Target different modes of action and manage multiple symptoms, for instance, an antiviral combination with an immune modulator to suppress the hyper-immune activity and simultaneously reduce viral load. 2) synergistic action between medications that target the same or different stages of the viral lifecycle or signalling pathways, allowing weak inhibitors to be more effective. 3) Enhance antiviral activity whilst lowering the risk of drug resistance. On the contrary, multiple combination therapies demerit safety concerns, including drug interactions, adverse drug reactions and multiple organ damage. For instance, combining baricitinib with remdesivir significantly suppresses virus load, infectivity, and hyper-inflammatory response. Kalil, Patterson et al209 administered a loading dose of 200 mg remdesivir IV on day 1 and 100 mg as a maintenance dose from day 2–10 or until they were discharged from the hospital. At the same time, 4 mg of oral baricitinib was administered to the COVID-19 patients daily for 14 days. The group of patients that received combination therapy exhibited a shorter mean time of recovery. Despite the concerns of secondary infections and thrombosis, supplementation of baricitinib did not increase the incidence of drug-drug interaction, adverse drug reactions and thromboembolic events.210

The COVID-19 pandemic has led to an urgent need to identify prognostic biomarkers and effective therapeutic approaches for severe-critical manifestations. Immune checkpoint molecules such as PD-1 and PD-L1 are crucial in regulating the immune response.211,212 According to Sabbatino, Conti et al213 serum PD-L1 levels have been found to have prognostic significance in COVID-19 patients, and its dysregulation is associated with COVID-19 pathogenesis. These findings suggest a potential role of the PD-1/PD-L1 axis in COVID-19 and support the need for clinical studies using PD-1/PD-L1 inhibitors.

Notably, the study from two Italian medical centres compared the clinical outcomes of COVID-19 in patients undergoing biological therapy targeting type 2 inflammation with those not receiving such therapies. The study concluded that monoclonal antibodies including omalizumab, mepolizumab, benralizumab, and dupilumab, targeting type 2 inflammation appeared safe, showing no worsening of symptoms, prolonged infection, or increased hospitalisations. However, an increase in common cold-like symptoms and dyspnoea during COVID-19 was reported. The findings suggest that anti-Th2 biological therapy should not be discontinued during SARS-CoV-2 infection. However, further controlled studies and analyses are needed to draw comprehensive conclusions about the safety and potential benefits of anti-type 2 mAbs in specific clinical contexts.214

Although modest dose and short-term (< 3 weeks) use of dexamethasone are considered safe, the risk of side effects increases with the dose and length of therapy noticeable.215 It exacerbates lymphopenia by further declining B- and T-cells in COVID-19 patients.216 Moreover, it is suggested that early corticosteroid (CS) usage may lead to greater viral loads and bolster virus replication.217 It is judicious to recall dexamethasone CYP3A4 inducing potential, ensuing interaction with the CYP3A4 substrates and decreasing plasma levels and efficacy of remdesivir in combination therapies.215 The secondary infections are potential sequelae of dexamethasone treatment as it exacerbates the risk of bacterial, invasive fungal and pre-existing infections like tuberculosis.218,219 Additionally, dexamethasone-induced strongyloidiasis and hyper-infection in low- and middle-income areas cannot be neglected.220

Recent studies have reported remdesivir adverse drug reactions ranging from minor flu-like symptoms to life-threatening end-stage hepatic failure in COVID-19 patients.221 According to Gupte, Hegde et al222 majority of the adverse events were reported nausea and elevated ALTs. Hepatic dysfunction and elevated transaminases were attributed to remdesivir hepatic biotransformation and hepatocyte permeability. Hepatocytes are inflicted by SARS-CoV-2 due to the prevalence of ACE2, which is later translated into elevated levels of ALT and bilirubin.223 Hence, it is difficult to diagnose whether transaminase abnormalities are caused by remdesivir or cytopathic injuries.224 Moreover, remdesivir’s high affinity for human mRNA polymerase is responsible for mitochondrial malfunction, exhibited in sinus bradycardia and QT prolongation in treated COVID-19 patients.225

A potent JAK inhibitor, baricitinib, has the potential to reactivate latent infections of herpes simplex, tuberculosis, hepatitis B, EBV, and herpes-zoster virus in COVID-19 patients.226,227 Deep vein thromboembolic events are frequently reported in rheumatoid arthritis patients as doses similarly administered for COVID-19 management.228 This is concerning as pulmonary embolism, and venous and arterial thrombotic events are commonly reported in COVID-19 patients.229 Kalil, Patterson, Mehta, Tomashek, Wolfe, Ghazaryan, Marconi, Ruiz-Palacios, Hsieh, Kline209 and the team terminated earlier due to grade three to four adverse drug reactions reported in 207 out of 507 (40.7%) participants. In this trial, a combination of baricitinib and remdesivir was administered, resulting in potentially life-threatening adverse effects, including hyperglycaemia, anaemia, lymphocytopenia, and acute kidney injury.

Concluding Remarks

As a result of the global spread of COVID-19, it is reasonable to assume that endocrine abnormalities will prevail over time. ACE2 and TMPRSS2 allow the SARS-CoV-2 to bind, gain entry, and impose cytopathic injuries on the host cells. Viral entry activates PRRs, NF-κB, JAK/STAT and MAPK pathways and significantly increases the production of cytokines, chemokines and interferons. IL-1β, IL-6, IFN-γ, and TNF-α are identified as key orchestrators of cytokine release syndrome. Where the cytolytic effect directly damages the host cells, indirect detrimental effects are inflected by the hosts’ hyper-immune response. The latter is characterised by SARS-CoV-2-induced cytokine storm, which leads to systemic inflammation and thyroid cell damage. The potential hormonal changes are due to the cytokine storm and due to which damage assessment during infection and after recovery from COVID-19 is highly warranted. The findings suggested that most patients are euthyroid and SAT and appear atypically without neck discomfort, but the development of AITD is significantly higher in COVID-19. New therapeutic regimens have been developed to cater specifically to COVID-19-associated thyroid dysregulations. Still, data from larger, randomised, and controlled trials are warranted to optimise the balance between the advantages and possible risks of these therapies. Additionally, it is crucial to determine the patient subgroups where this ratio is most likely to promote a safe and effective treatment outcome.

It is rational to shift the focus from monotherapy to drug combinations, as there is no gold standard treatment, and several repurposed drugs have failed to demonstrate meaningful value in recent clinical trials. Further randomised controlled studies might improve overall therapy effectiveness by emphasising the safety and efficacy of the combined therapies against COVID-19-related thyroid diseases. Furthermore, interventions targeting various stages of disease processes, including viral replication, cytokine-induced damage and thromboembolic events, should be the vision and mission of future studies. Combination therapies delivered at the right time can effectively address various signalling pathways and attenuate disease progression. In contrast, some drug combination therapies have shown promising effects in small groups or secluded clinical settings. The disease’s intricacy demands us to look into combining independent treatments to improve their potential in a much larger population.

Funding Statement

This work was supported by the Deanship of Scientific Research, Vice Presidency for Graduate Studies and Scientific Research, King Faisal University, Saudi Arabia [KFU241533].

Abbreviation

AAK, AP2-associated protein kinase; ADCC, antibody-dependent cytotoxicity; AITD, autoimmune thyroid disorder: Akt, protein kinase B; ALR, absent in melanoma-2-like receptor; ARDS, acute respiratory distress syndrome; ACE, angiotensin-converting enzyme-2; ALT, alanine transaminase; APC, antigen-presenting cells; AST, aspartate aminotransferase; AT, angiotensin; CARD, caspase recruitment domain; CCL, C-C motif chemokine ligand; CLR, C-type lectin receptor; Cmax, maximum serum concentration; CoV, coronavirus; COVID-19, coronavirus Disease 2019; CREB, cAMP response element-binding protein; CRP, C-reactive protein; CRS, cytokine release syndrome; CTD, C-terminal Domain; DAMP, damage-associated molecular pattern; eNOS, endothelial nitric oxide synthase; ERGIC, endoplasmic reticulum-golgi intermediate compartment; ERK, extracellular signal-regulated kinase; ESS, euthyroid sick syndrome; GAG, glycosaminoglycan; GAK, G-associated kinase; GD, graves’ disease; GPCR, G-protein coupled receptor; GR, glucocorticoid receptor; GSH, glutathione; HepG2, human hepatoma cell line; HLA, human leukocyte antigen; HPT, hypothalamic-pituitary-thyroid; HT, hashimoto thyroiditis; ICAM, intracellular adhesion molecule, IFN, interferon; Ig, immunoglobulin; IκK, inhibitors of NF-κB Kinase; IL, interleukin; IL-6 Amp, interleukin-6 amplifier; iNOS, inducible nitric oxide synthase; ISG, interferon-stimulated gene; JAK, janus kinase; JNK, C-jun N-terminal kinase; MAPK, mitogen-activated protein kinase; MERS-CoV, middle east respiratory syndrome coronavirus; MHC, major histocompatibility complex; MKP, MAPK phosphatase; MYD88, myeloid differentiation primary response 88; NAC, N-acetyl-cysteine; NADPH, nicotine adenine dinucleotide phosphate; NF-κB, Nuclear Factor Kappa-light-chain-enhancer of activated B cells; NIS, sodium iodide symporter; NK, natural killer cell; NLR, nucleotide oligomerization domain-like receptor; NLRP3, NLR pyrin domain containing 3; NSP, non-structural protein; NTD, N-terminal domain; PAMP, pathogen-associated molecular pattern; PDK, phosphoinositide-dependent protein kinase; PG, prostaglandin; PI3K, phosphatidylinositol 3,4,5 triphosphate kinase; PRR, pattern-recognition receptor; PVN, paraventricular nucleus; RAS, renin-angiotensin system; RdRp, RNA-dependent RNA polymerase; RNS, reactive nitrogen species; ROS, reactive oxygen species; RBD, receptor-binding domain; RER, rough endoplasmic reticulum; RLR, retinoic acid-inducible gene-1-like Receptor; RORγt, RAR-related orphan receptor-γ; rT3, reverse triiodothyronine; SARS, severe acute respiratory syndrome; SARS-CoV-2, severe acute respiratory syndrome coronavirus-2; SAT, subacute thyroiditis; STAT, signal transducer and activator of transcription; TAC, total antioxidant capacity; TAK, transforming-growth-factor-β-activated kinase; TBG, thyroxine binding globulin; TCR, T-cell receptor; Tg, thyroglobulin; Th, T-helper; TLR, toll-like receptor; TMPRSS2, transmembrane protease serine protease 2; TNF, tumor necrosis factor; TPO, thyroid peroxidase; TRAb, TSH receptor autoantibodies; TRAF, TNF receptor-associated factor; TRE, thyroid hormone response element; Treg, regulatory T-cell; TSH, thyroid-stimulating hormone; TYK, tyrosine kinase; T3, triiodothyronine; T4, thyroxine; ULN, upper limit of normal; VCAM, vascular cell adhesion molecules; WHO, World Health Organization.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Disclosure

The authors report no conflict of interest for the present paper.

References

  • 1.Huang C, Wang Y, Li X, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506. doi: 10.1016/S0140-6736(20)30183-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gupta A, Madhavan MV, Sehgal K, et al. Extrapulmonary manifestations of COVID-19. Nat Med. 2020;26(7):1017–1032. doi: 10.1038/s41591-020-0968-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lee I-C, Huo T-I, Huang Y-H. Gastrointestinal and liver manifestations in patients with COVID-19. J Chin Med Assoc. 2020;83(6):521–523. doi: 10.1097/JCMA.0000000000000319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Li M-Y, Li L, Zhang Y, Wang X-S. Expression of the SARS-CoV-2 cell receptor gene ACE2 in a wide variety of human tissues. Infect Diseases Poverty. 2020;9(02):23–29. doi: 10.1186/s40249-020-00662-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Yan R, Zhang Y, Li Y, Xia L, Guo Y, Zhou Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science. 2020;367(6485):1444–1448. doi: 10.1126/science.abb2762 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Zhao P, Praissman JL, Grant OC, et al. Virus-receptor interactions of glycosylated SARS-CoV-2 spike and human ACE2 receptor. Cell Host Microbe. 2020;28(4):586–601.e586. doi: 10.1016/j.chom.2020.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Mattar SAM, Koh SJQ, Rama Chandran S, Cherng BPZ. Subacute thyroiditis associated with COVID-19. BMJ Case Rep. 2020;13(8):e237336. doi: 10.1136/bcr-2020-237336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sohrabpour S, Heidari F, Karimi E, Ansari R, Tajdini A, Heidari F. Subacute thyroiditis in COVID-19 patients. Europ Thyr J. 2021;9(6):321–323. doi: 10.1159/000511707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Brancatella A, Viola N, Santini F, Latrofa F. COVID-induced thyroid autoimmunity. Best Pract Res Clin Endocrinol Metab. 2023;37(2):101742. doi: 10.1016/j.beem.2023.101742 [DOI] [PubMed] [Google Scholar]
  • 10.Lui DT, Lee CH, Chow WS, et al. Insights from prospective follow-up of thyroid function and autoimmunity among covid-19 survivors. J Endoc Soc. 2021;5(Supplement_1):A840–A841. doi: 10.1210/jendso/bvab048.1715 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wang W, Su X, Ding Y, et al. Thyroid function abnormalities in COVID-19 patients. Front Endocrinol. 2021;11:623792. doi: 10.3389/fendo.2020.623792 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zhang Y, Lin F, Tu W, et al. Thyroid dysfunction may be associated with poor outcomes in patients with COVID-19. Molecul Cell Endocrinol. 2021;521:111097. doi: 10.1016/j.mce.2020.111097 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Brancatella A, Ricci D, Viola N, Sgrò D, Santini F, Latrofa F. Subacute thyroiditis after Sars-COV-2 infection. J Clin Endocrinol Metab. 2020;105(7):2367–2370. doi: 10.1210/clinem/dgaa276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Giovanella L, Ruggeri RM, Ovčariček PP, Campenni A, Treglia G, Deandreis D. Prevalence of thyroid dysfunction in patients with COVID-19: a systematic review. Clin Transl Imaging. 2021;9(3):233–240. doi: 10.1007/s40336-021-00419-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Scappaticcio L, Pitoia F, Esposito K, Piccardo A, Trimboli P. Impact of COVID-19 on the thyroid gland: an update. Rev Endocr Metab Disord. 2021;22(4):803–815. doi: 10.1007/s11154-020-09615-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Aslani M, Mortazavi-Jahromi SS, Mirshafiey A. Cytokine storm in the pathophysiology of COVID-19: possible functional disturbances of miRNAs. Int Immunopharmacol. 2021;101:108172. doi: 10.1016/j.intimp.2021.108172 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Speer G, Somogyi P. Thyroid complications of SARS and coronavirus disease 2019 (COVID-19). Endocr J. 2021;68(2):129–136. doi: 10.1507/endocrj.EJ20-0443 [DOI] [PubMed] [Google Scholar]
  • 18.Feghali K, Atallah J, Norman C. Manifestations of thyroid disease post COVID-19 illness: report of Hashimoto thyroiditis, Graves’ disease, and subacute thyroiditis. J Clin Translat Endocrinol. 2021;22:100094. doi: 10.1016/j.jecr.2021.100094 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Clausen CL, Rasmussen ÅK, Johannsen TH, et al. Thyroid function in COVID-19 and the association with cytokine levels and mortality. Endocr Connections. 2021;10(10):1234–1242. doi: 10.1530/EC-21-0301 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lania A, Sandri MT, Cellini M, Mirani M, Lavezzi E, Mazziotti G. Thyrotoxicosis in patients with COVID-19: the THYRCOV study. Europ J Endocrinol. 2020;183(4):381–387. doi: 10.1530/EJE-20-0335 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ashrafi S, Hatami H, Bidhendi-Yarandi R, Panahi MH. The prevalence of thyroid disorders in COVID-19 patients: a systematic review and meta-analysis. BMC Endocr Disord. 2024;24(1):5. doi: 10.1186/s12902-023-01534-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Barrera EC, Cisneros TA, Fuentes MSD. Subacute Thyroiditis Associated with COVID-19; 2021.
  • 23.Bellastella G, Maiorino M, Esposito K. Endocrine complications of COVID-19: what happens to the thyroid and adrenal glands? J Endocrinol Invest. 2020;43:1169–1170. doi: 10.1007/s40618-020-01311-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Damara FA, Muchamad GR, Ikhsani R, Syafiyah AH, Bashari MH, Bashari MH. Thyroid disease and hypothyroidism are associated with poor COVID-19 outcomes: a systematic review, meta-analysis, and meta-regression. Diabetes Metab Syndr. 2021;15(6):102312. doi: 10.1016/j.dsx.2021.102312 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Khatri A, Charlap E, Kim A. Subacute thyroiditis from COVID-19 infection: a case report and review of literature. Europ Thyr J. 2021;9(6):324–328. doi: 10.1159/000511872 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Van Gerwen M, Alsen M, Little C, et al. Outcomes of patients with hypothyroidism and COVID-19: a retrospective cohort study. Front Endocrinol. 2020;11:565. doi: 10.3389/fendo.2020.00565 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Rossetti CL, Cazarin J, Hecht F, et al. COVID-19 and thyroid function: what do we know so far? Front Endocrinol. 2022;13:1041676. doi: 10.3389/fendo.2022.1041676 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Qutob RA, Alhusaini BA, Alzmamy SA, et al. Knowledge and awareness about risk factors, clinical manifestations, and prevention of thyroid disorders in the era of COVID-19 and their association with socioeconomic status among the general population in Riyadh, Saudi Arabia. Cureus. 2023;15(11):e48878. doi: 10.7759/cureus.48878 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Şendur SN, Oğuz SH, Ünlütürk U. COVID-19 vaccination and thyroiditis. Best Pract Res Clin Endocrinol Metab. 2023;37(4):101759. doi: 10.1016/j.beem.2023.101759 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bosch BJ, Van der Zee R, De Haan CA, Rottier PJ. The coronavirus spike protein is a class I virus fusion protein: structural and functional characterization of the fusion core complex. J Virol. 2003;77(16):8801–8811. doi: 10.1128/JVI.77.16.8801-8811.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Hu B, Guo H, Zhou P, Shi Z-L. Characteristics of SARS-CoV-2 and COVID-19. Nat Rev Microbiol. 2021;19(3):141–154. doi: 10.1038/s41579-020-00459-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yuki K, Fujiogi M, Koutsogiannaki S. COVID-19 pathophysiology: a review. Clin Immunol. 2020;215:108427. doi: 10.1016/j.clim.2020.108427 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chen Y, Liu Q, Guo D. Emerging coronaviruses: genome structure, replication, and pathogenesis. J Med Virol. 2020;92(4):418–423. doi: 10.1002/jmv.25681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fehr AR, Perlman S. Coronaviruses: an overview of their replication and pathogenesis. Coronaviruses. 2015;2015:1–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Lan J, Ge J, Yu J, et al. Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature. 2020;581(7807):215–220. doi: 10.1038/s41586-020-2180-5 [DOI] [PubMed] [Google Scholar]
  • 36.Shang J, Wan Y, Luo C, et al. Cell entry mechanisms of SARS-CoV-2. Proc Natl Acad Sci. 2020;117(21):11727–11734. doi: 10.1073/pnas.2003138117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sungnak W, Huang N, Bécavin C, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat Med. 2020;26(5):681–687. doi: 10.1038/s41591-020-0868-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dhama K, Khan S, Tiwari R, et al. Coronavirus disease 2019–COVID-19. Clin Microbiol Rev. 2020;33(4):e00028–00020. doi: 10.1128/CMR.00028-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Samudrala PK, Kumar P, Choudhary K, et al. Virology, pathogenesis, diagnosis and in-line treatment of COVID-19. Eur J Pharmacol. 2020;883:173375. doi: 10.1016/j.ejphar.2020.173375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zhou P, Yang X-L, Wang X-G, et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270–273. doi: 10.1038/s41586-020-2012-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Knoops K, Kikkert M, SHvd W, et al. SARS-coronavirus replication is supported by a reticulovesicular network of modified endoplasmic reticulum. PLoS Biol. 2008;6(9):e226. doi: 10.1371/journal.pbio.0060226 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chams N, Chams S, Badran R, et al. COVID-19: a multidisciplinary review. Front Public Health. 2020;8:383. doi: 10.3389/fpubh.2020.00383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kuba K, Imai Y, Rao S, et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus–induced lung injury. Nat Med. 2005;11(8):875–879. doi: 10.1038/nm1267 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Imai Y, Kuba K, Rao S, et al. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005;436(7047):112–116. doi: 10.1038/nature03712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Attiq A, Yao LJ, Afzal S, Khan MA. The triumvirate of NF-κB, inflammation and cytokine storm in COVID-19. Int Immunopharmacol. 2021;101:108255. doi: 10.1016/j.intimp.2021.108255 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zheng KI, Feng G, Liu WY, Targher G, Byrne CD, Zheng MH. Extrapulmonary complications of COVID‐19: a multisystem disease? J Med Virol. 2021;93(1):323–335. doi: 10.1002/jmv.26294 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mateu-Salat M, Urgell E, Chico A. SARS-COV-2 as a trigger for autoimmune disease: report of two cases of Graves’ disease after COVID-19. J Endocrinol Invest. 2020;43(10):1527–1528. doi: 10.1007/s40618-020-01366-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Caron P Thyroid disorders and SARS-CoV-2 infection: from pathophysiological mechanism to patient management. Paper presented at: Annales D’endocrinologie; 2020. [DOI] [PMC free article] [PubMed]
  • 49.Li X, Geng M, Peng Y, Meng L, Lu S. Molecular immune pathogenesis and diagnosis of COVID-19. J Pharm Anal. 2020;10(2):102–108. doi: 10.1016/j.jpha.2020.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Gu J, Gong E, Zhang B, et al. Multiple organ infection and the pathogenesis of SARS. J Exp Med. 2005;202(3):415–424. doi: 10.1084/jem.20050828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wei L, Sun S, Zhang J, et al. Endocrine cells of the adenohypophysis in severe acute respiratory syndrome (SARS). Biochem Cell Biol. 2010;88(4):723–730. doi: 10.1139/O10-022 [DOI] [PubMed] [Google Scholar]
  • 52.Chen M, Zhou W, Xu W. Thyroid function analysis in 50 patients with COVID-19: a retrospective study. Thyroid. 2021;31(1):8–11. doi: 10.1089/thy.2020.0363 [DOI] [PubMed] [Google Scholar]
  • 53.Amarante-Mendes GP, Adjemian S, Branco LM, Zanetti LC, Weinlich R, Bortoluci KR. Pattern recognition receptors and the host cell death molecular machinery. Front Immunol. 2018;9:2379. doi: 10.3389/fimmu.2018.02379 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Li D, Wu M, Bi Z. Pattern recognition receptors in health and diseases. Signal Transduct Targ Ther. 2021;6(1):1–24. doi: 10.1038/s41392-020-00451-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.He S, He J, Chen J, Chen F, Ou S. Research advancement of innate immunity and pattern recognition receptors. ChinJ Animal Nutr. 2017;29(11):3844–3851. [Google Scholar]
  • 56.Onomoto K, Onoguchi K, Yoneyama M. Regulation of RIG-I-like receptor-mediated signaling: interaction between host and viral factors. Cell Mol Immunol. 2021;18(3):539–555. doi: 10.1038/s41423-020-00602-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yoneyama M, Onomoto K, Jogi M, Akaboshi T, Fujita T. Viral RNA detection by RIG-I-like receptors. Curr Opin Immunol. 2015;32:48–53. doi: 10.1016/j.coi.2014.12.012 [DOI] [PubMed] [Google Scholar]
  • 58.Medzhitov R, Preston-Hurlburt P, Janeway CA. A human homologue of the Drosophila Toll protein signals activation of adaptive immunity. Nature. 1997;388(6640):394–397. doi: 10.1038/41131 [DOI] [PubMed] [Google Scholar]
  • 59.Kumar V. Toll-like receptors in sepsis-associated cytokine storm and their endogenous negative regulators as future immunomodulatory targets. Int Immunopharmacol. 2020;89:107087. doi: 10.1016/j.intimp.2020.107087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Giuliani C, Bucci I, Napolitano G. The role of the transcription factor nuclear factor-kappa B in thyroid autoimmunity and cancer. Front Endocrinol. 2018;9:471. doi: 10.3389/fendo.2018.00471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Harii N, Lewis CJ, Vasko V, et al. Thyrocytes express a functional toll-like receptor 3: overexpression can be induced by viral infection and reversed by phenylmethimazole and is associated with Hashimoto’s autoimmune thyroiditis. Mol Endocrinol. 2005;19(5):1231–1250. doi: 10.1210/me.2004-0100 [DOI] [PubMed] [Google Scholar]
  • 62.Ferrari SM, Fallahi P, Elia G, et al. Thyroid autoimmune disorders and cancer. Paper presented at: Seminars in cancer biology 2020. [DOI] [PubMed]
  • 63.Yao LJ, Jalil J, Attiq A, Hui CC, Zakaria NA. The medicinal uses, toxicities and anti-inflammatory activity of Polyalthia species (Annonaceae). J Ethnopharmacol. 2019;229:303–325. doi: 10.1016/j.jep.2018.10.001 [DOI] [PubMed] [Google Scholar]
  • 64.Jalil J, Attiq A, Hui CC, Yao LJ, Zakaria NA. Modulation of inflammatory pathways, medicinal uses and toxicities of Uvaria species: potential role in the prevention and treatment of inflammation. Inflammopharmacology. 2020;28(5):1195–1218. doi: 10.1007/s10787-020-00734-2 [DOI] [PubMed] [Google Scholar]
  • 65.Attiq A, Jalil J, Husain K. Annonaceae: breaking the wall of inflammation. Front Pharmacol. 2017;8:752. doi: 10.3389/fphar.2017.00752 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hariharan A, Hakeem AR, Radhakrishnan S, Reddy MS, Rela M. The role and therapeutic potential of NF-kappa-B pathway in severe COVID-19 patients. Inflammopharmacology. 2021;29(1):91–100. doi: 10.1007/s10787-020-00773-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kwakkel J, Wiersinga WM, Boelen A. Differential involvement of nuclear factor-κB and activator protein-1 pathways in the interleukin-1β-mediated decrease of deiodinase type 1 and thyroid hormone receptor β1 mRNA. J Endocrinol. 2006;189(1):37–44. doi: 10.1677/joe.1.06354 [DOI] [PubMed] [Google Scholar]
  • 68.Castro G, Liu X, Ngo K, et al. RORγt and RORα signature genes in human Th17 cells. PLoS One. 2017;12(8):e0181868. doi: 10.1371/journal.pone.0181868 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Hou W, Kang HS, Kim BS. Th17 cells enhance viral persistence and inhibit T cell cytotoxicity in a model of chronic virus infection. J Exp Med. 2009;206(2):313–328. doi: 10.1084/jem.20082030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Josefowicz SZ, Lu L-F, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol. 2012;30:531. doi: 10.1146/annurev.immunol.25.022106.141623 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Attiq A, Afzal S. Trinity of inflammation, innate immune cells and cross-talk of signalling pathways in tumour microenvironment. Front Pharmacol. 2023;2023:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Long M, Park S-G, Strickland I, Hayden MS, Ghosh S. Nuclear factor-κB modulates regulatory T cell development by directly regulating expression of Foxp3 transcription factor. Immunity. 2009;31(6):921–931. doi: 10.1016/j.immuni.2009.09.022 [DOI] [PubMed] [Google Scholar]
  • 73.Yamaoka K, Saharinen P, Pesu M, Holt VE, Silvennoinen O, O’Shea JJ. The janus kinases (jaks). Genome Biol. 2004;5(12):1–6. doi: 10.1186/gb-2004-5-12-253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wu D, Yang XO. TH17 responses in cytokine storm of COVID-19: an emerging target of JAK2 inhibitor Fedratinib. J Microbiol Immunol Infect. 2020;53(3):368–370. doi: 10.1016/j.jmii.2020.03.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Pfitzner E, Kliem S, Baus D, Litterst M. The role of STATs in inflammation and inflammatory diseases. Curr Pharm Des. 2004;10(23):2839–2850. doi: 10.2174/1381612043383638 [DOI] [PubMed] [Google Scholar]
  • 76.de Prati AC, Ciampa AR, Cavalieri E, et al. STAT1 as a new molecular target of anti-inflammatory treatment. Curr Med Chem. 2005;12(16):1819–1828. doi: 10.2174/0929867054546645 [DOI] [PubMed] [Google Scholar]
  • 77.Decker T, Stockinger S, Karaghiosoff M, Müller M, Kovarik P. IFNs and STATs in innate immunity to microorganisms. J Clin Invest. 2002;109(10):1271–1277. doi: 10.1172/JCI0215770 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Grifoni A, Weiskopf D, Ramirez SI, et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. 2020;181(7):1489–1501.e1415. doi: 10.1016/j.cell.2020.05.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kisseleva T, Bhattacharya S, Braunstein J, Schindler C. Signaling through the JAK/STAT pathway, recent advances and future challenges. Gene. 2002;285(1–2):1–24. doi: 10.1016/s0378-1119(02)00398-0 [DOI] [PubMed] [Google Scholar]
  • 80.Sosonkina N, Starenki D, Park J-I. The role of STAT3 in thyroid cancer. Cancers (Basel). 2014;6(1):526–544. doi: 10.3390/cancers6010526 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Nishihara M, Ogura H, Ueda N, et al. IL-6–gp130–STAT3 in T cells directs the development of IL-17+ Th with a minimum effect on that of Treg in the steady state. Int Immunol. 2007;19(6):695–702. doi: 10.1093/intimm/dxm045 [DOI] [PubMed] [Google Scholar]
  • 82.Kotkowska A, Sewerynek E, Domańska D, Pastuszak-Lewandoska D, Brzeziańska E. Single nucleotide polymorphisms in the STAT3 gene influence AITD susceptibility, thyroid autoantibody levels, and IL6 and IL17 secretion. Cell. Mol. Biol. Lett. 2015;20(1):88–101. doi: 10.1515/cmble-2015-0004 [DOI] [PubMed] [Google Scholar]
  • 83.Murakami M, Hirano T. A four-step model for the IL-6 amplifier, a regulator of chronic inflammations in tissue-specific MHC class II-associated autoimmune diseases. Front Immunol. 2011;2:22. doi: 10.3389/fimmu.2011.00022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lee J, Nakagiri T, Oto T, et al. IL-6 amplifier, NF-κB–triggered positive feedback for IL-6 signaling, in grafts is involved in allogeneic rejection responses. J Immunol. 2012;189(4):1928–1936. doi: 10.4049/jimmunol.1103613 [DOI] [PubMed] [Google Scholar]
  • 85.Cooke KR, Coghill JM, Serody JS. Chemokines and graft-versus-host disease. In: Immune Biology of Allogeneic Hematopoietic Stem Cell Transplantation. Elsevier; 2019:323–347. [Google Scholar]
  • 86.Thalhamer T, McGrath M, Harnett M. MAPKs and their relevance to arthritis and inflammation. Rheumatology. 2008;47(4):409–414. doi: 10.1093/rheumatology/kem297 [DOI] [PubMed] [Google Scholar]
  • 87.Noubade R, Krementsov DN, Del Rio R, et al. Activation of p38 MAPK in CD4 T cells controls IL-17 production and autoimmune encephalomyelitis. Blood J Am Soc Hematol. 2011;118(12):3290–3300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wajner SM, Goemann IM, Bueno AL, Larsen PR, Maia AL. IL-6 promotes nonthyroidal illness syndrome by blocking thyroxine activation while promoting thyroid hormone inactivation in human cells. J Clin Invest. 2011;121(5):1834–1845. doi: 10.1172/JCI44678 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Romitti M, Wajner SM, Ceolin L, et al. MAPK and SHH pathways modulate type 3 deiodinase expression in papillary thyroid carcinoma. Endocr Relat Cancer. 2016;23(3):135–146. doi: 10.1530/ERC-15-0162 [DOI] [PubMed] [Google Scholar]
  • 90.Sharma R, Traore K, Trush M, Rose N, Burek CL. Intracellular adhesion molecule-1 up-regulation on thyrocytes by iodine of non-obese diabetic. H2h4 mice is reactive oxygen species-dependent. Clin Exp Immunol. 2008;152(1):13–20. doi: 10.1111/j.1365-2249.2008.03590.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Afzal S, Abdul Manap AS, Attiq A, Albokhadaim I, Kandeel M, Alhojaily SM. From imbalance to impairment: the central role of reactive oxygen species in oxidative stress-induced disorders and therapeutic exploration. Front Pharmacol. 2023;2023:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Duntas LH. The catalytic role of iodine excess in loss of homeostasis in autoimmune thyroiditis. Curr Opin Endocrino. 2018;25(5):347–352. doi: 10.1097/MED.0000000000000425 [DOI] [PubMed] [Google Scholar]
  • 93.Hong W, Yang J, Bi Z, He C, Lei H, Yu W. A mouse model for SARS-CoV-2-induced acute respiratory distress syndrome. Signal Transduct Targ Ther. 2021;6(1):1–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zhang C, Wu Z, Li J-W, Zhao H, Wang G-Q. Cytokine release syndrome in severe COVID-19: interleukin-6 receptor antagonist tocilizumab may be the key to reduce mortality. Int J Antimicrob Agents. 2020;55(5):105954. doi: 10.1016/j.ijantimicag.2020.105954 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Croce L, Gangemi D, Ancona G, et al. The cytokine storm and thyroid hormone changes in COVID-19. J Endocrinol Invest. 2021;44(5):891–904. doi: 10.1007/s40618-021-01506-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Jimenez C, Moran SA, Sereti I, et al. Graves’ disease after interleukin-2 therapy in a patient with human immunodeficiency virus infection. Thyroid. 2004;14(12):1097–1102. doi: 10.1089/thy.2004.14.1097 [DOI] [PubMed] [Google Scholar]
  • 97.Moon B-G, Takaki S, Miyake K, Takatsu K. The role of IL-5 for mature B-1 cells in homeostatic proliferation, cell survival, and Ig production. J Immunol. 2004;172(10):6020–6029. doi: 10.4049/jimmunol.172.10.6020 [DOI] [PubMed] [Google Scholar]
  • 98.Wajner SM, Maia AL. New insights toward the acute non-thyroidal illness syndrome. Front Endocrinol. 2012;3:8. doi: 10.3389/fendo.2012.00008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Soehnlein O, Lindbom L, Weber C. Mechanisms underlying neutrophil-mediated monocyte recruitment. Blood J Am Soc Hematol. 2009;114(21):4613–4623. [DOI] [PubMed] [Google Scholar]
  • 100.Mannino MH, Zhu Z, Xiao H, Bai Q, Wakefield MR, Fang Y. The paradoxical role of IL-10 in immunity and cancer. Cancer Lett. 2015;367(2):103–107. doi: 10.1016/j.canlet.2015.07.009 [DOI] [PubMed] [Google Scholar]
  • 101.Nagayama Y, Mizuguchi H, Hayakawa T, Niwa M, McLachlan SM, Rapoport B. Prevention of autoantibody-mediated Graves’-like hyperthyroidism in mice with IL-4, a Th2 cytokine. J Immunol. 2003;170(7):3522–3527. doi: 10.4049/jimmunol.170.7.3522 [DOI] [PubMed] [Google Scholar]
  • 102.Moens L, Tangye SG. Cytokine-mediated regulation of plasma cell generation: IL-21 takes center stage. Front Immunol. 2014;5:65. doi: 10.3389/fimmu.2014.00065 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Mezosi E, Wang SH, Utsugi S, et al. Interleukin-1beta and tumor necrosis factor (TNF)-alpha sensitize human thyroid epithelial cells to TNF-related apoptosis-inducing ligand-induced apoptosis through increases in procaspase-7 and Bid, and the down-regulation of p44/42 mitogen-activated protein kinase activity. J Clin Endocrinol Metab. 2004;89(1):250–257. doi: 10.1210/jc.2003-030697 [DOI] [PubMed] [Google Scholar]
  • 104.Antonelli A, Ferrari SM, Frascerra S, et al. Increase of circulating CXCL9 and CXCL11 associated with euthyroid or subclinically hypothyroid autoimmune thyroiditis. J Clin Endocrinol Metab. 2011;96(6):1859–1863. doi: 10.1210/jc.2010-2905 [DOI] [PubMed] [Google Scholar]
  • 105.Gaffen SL, Jain R, Garg AV, Cua DJ. The IL-23–IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol. 2014;14(9):585–600. doi: 10.1038/nri3707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kawashima A, Tanigawa K, Akama T, Yoshihara A, Ishii N, Suzuki K. Innate immune activation and thyroid autoimmunity. J Clin Endocrinol Metab. 2011;96(12):3661–3671. doi: 10.1210/jc.2011-1568 [DOI] [PubMed] [Google Scholar]
  • 107.Marazuela M, Garcia-Lopez MA, Figueroa-Vega N, et al. Regulatory T cells in human autoimmune thyroid disease. J Clin Endocrinol Metab. 2006;91(9):3639–3646. doi: 10.1210/jc.2005-2337 [DOI] [PubMed] [Google Scholar]
  • 108.Bhavsar I, Miller CS, Al-Sabbagh M. Macrophage inflammatory protein-1 alpha (MIP-1 alpha)/CCL3: as a biomarker. Gen Meth Biomark Res Their Applic. 2015;2015:223. [Google Scholar]
  • 109.Rotondi M, Chiovato L, Romagnani S, Serio M, Romagnani P. Role of chemokines in endocrine autoimmune diseases. Endocr Rev. 2007;28(5):492–520. doi: 10.1210/er.2006-0044 [DOI] [PubMed] [Google Scholar]
  • 110.Schutyser E, Struyf S, Van Damme J. The CC chemokine CCL20 and its receptor CCR6. Cytokine Growth Factor Rev. 2003;14(5):409–426. doi: 10.1016/S1359-6101(03)00049-2 [DOI] [PubMed] [Google Scholar]
  • 111.Rasmussen K, Bendtzen K, Feldt-Rasmussen U. Thyrocyte-interleukin-1 interactions. Exp Clin Endocrinol Diabetes. 2000;108(02):67–71. doi: 10.1055/s-2000-5797 [DOI] [PubMed] [Google Scholar]
  • 112.Malik A, Kanneganti TD. Function and regulation of IL‐1α in inflammatory diseases and cancer. Immunol Rev. 2018;281(1):124–137. doi: 10.1111/imr.12615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Giordano C, Stassi G, De Maria R, et al. Potential involvement of Fas and its ligand in the pathogenesis of Hashimoto’s thyroiditis. Science. 1997;275:5302):960–963. doi: 10.1126/science.275.5302.960 [DOI] [PubMed] [Google Scholar]
  • 114.Brenner S, Miller JH. Brenner’s Encyclopedia of Genetics. Elsevier Science; 2014. [Google Scholar]
  • 115.Yang S, Wang J, Brand DD, Zheng SG. Role of TNF–TNF receptor 2 signal in regulatory T cells and its therapeutic implications. Front Immunol. 2018;9:784. doi: 10.3389/fimmu.2018.00784 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Lee HJ, Lombardi A, Stefan M, et al. Cd40 signaling in graves disease is mediated through canonical and noncanonical thyroidal nuclear factor κ b activation. Endocrinology. 2017;158(2):410–418. doi: 10.1210/en.2016-1609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Jacobson E, Huber A, Akeno N, et al. A CD40 Kozak sequence polymorphism and susceptibility to antibody-mediated autoimmune conditions: the role of CD40 tissue-specific expression. Genes Immun. 2007;8(3):205–214. doi: 10.1038/sj.gene.6364375 [DOI] [PubMed] [Google Scholar]
  • 118.Ferrari SM, Ruffilli I, Elia G, et al. Chemokines in hyperthyroidism. J Clin Translat Endocrinol. 2019;16:100196. doi: 10.1016/j.jcte.2019.100196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Boye K, Billottet C, Pujol N, Alves I, Bikfalvi A. Ligand activation induces different conformational changes in CXCR3 receptor isoforms as evidenced by plasmon waveguide resonance (PWR). Sci Rep. 2017;7(1):1–11. doi: 10.1038/s41598-017-11151-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Coperchini F, Chiovato L, Rotondi M. Interleukin-6, CXCL10 and infiltrating macrophages in COVID-19-related cytokine storm: not one for all but all for one! Front Immunol. 2021;12:668507. doi: 10.3389/fimmu.2021.668507 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Dembic Z. The Cytokines of the Immune System: The Role of Cytokines in Disease Related to Immune Response. Academic Press; 2015. [Google Scholar]
  • 122.Docter R, Krenning E, De Jong M, Hennemann G. The sick euthyroid syndrome: changes in thyroid hormone serum parameters and hormone metabolism. Clin Endocrinol. 1993;39(5):499–518. doi: 10.1111/j.1365-2265.1993.tb02401.x [DOI] [PubMed] [Google Scholar]
  • 123.de Vries EM, Fliers E, Boelen A. The molecular basis of the non-thyroidal illness syndrome. J Endocrinol. 2015;225(3):R67–81. doi: 10.1530/JOE-15-0133 [DOI] [PubMed] [Google Scholar]
  • 124.Zou R, Wu C, Zhang S, et al. Euthyroid sick syndrome in patients with COVID-19. Front Endocrinol. 2020;11:566439. doi: 10.3389/fendo.2020.566439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Takahashi-Tezuka M, Yoshida Y, Fukada T, et al. Gab1 acts as an adapter molecule linking the cytokine receptor gp130 to ERK mitogen-activated protein kinase. Mol Cell Biol. 1998;18(7):4109–4117. doi: 10.1128/MCB.18.7.4109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Kester MH, Kuiper GG, Versteeg R, Visser TJ. Regulation of type III iodothyronine deiodinase expression in human cell lines. Endocrinology. 2006;147(12):5845–5854. doi: 10.1210/en.2006-0590 [DOI] [PubMed] [Google Scholar]
  • 127.Baqui M, Botero D, Gereben B, et al. Human type 3 iodothyronine selenodeiodinase is located in the plasma membrane and undergoes rapid internalization to endosomes. J Biol Chem. 2003;278(2):1206–1211. doi: 10.1074/jbc.M210266200 [DOI] [PubMed] [Google Scholar]
  • 128.Schafer FQ, Buettner GR. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med. 2001;30(11):1191–1212. doi: 10.1016/S0891-5849(01)00480-4 [DOI] [PubMed] [Google Scholar]
  • 129.Attiq A, Jalil J, Husain K, Mohamad HF, Ahmad A. Luteolin and apigenin derived glycosides from Alphonsea elliptica abrogate LPS-induced inflammatory responses in human plasma. J Ethnopharmacol. 2021;275:114120. doi: 10.1016/j.jep.2021.114120 [DOI] [PubMed] [Google Scholar]
  • 130.Attiq A, Jalil J, Husain K, Jamal JA, Ismail EN. A new prenylated benzoquinone from Cyathocalyx pruniferus abrogates LPS-induced inflammatory responses associated with PGE2, COX-2 and cytokines biosynthesis in human plasma. Inflammopharmacology. 2021;29(3):841–854. doi: 10.1007/s10787-021-00807-w [DOI] [PubMed] [Google Scholar]
  • 131.Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J. Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol. 2007;39(1):44–84. doi: 10.1016/j.biocel.2006.07.001 [DOI] [PubMed] [Google Scholar]
  • 132.Darras VM, Van Herck SL. Iodothyronine deiodinase structure and function: from ascidians to humans. J Endocrinol. 2012;215(2):189–206. doi: 10.1530/JOE-12-0204 [DOI] [PubMed] [Google Scholar]
  • 133.Kuiper GG, Klootwijk W, Visser TJ. Substitution of cysteine for a conserved alanine residue in the catalytic center of type II iodothyronine deiodinase alters interaction with reducing cofactor. Endocrinology. 2002;143(4):1190–1198. doi: 10.1210/endo.143.4.8738 [DOI] [PubMed] [Google Scholar]
  • 134.Chen I-Y, Moriyama M, Chang M-F, Ichinohe T. Severe acute respiratory syndrome coronavirus viroporin 3a activates the NLRP3 inflammasome. Front Microbiol. 2019;10:50. doi: 10.3389/fmicb.2019.00050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Brigelius-Flohé R, Banning A, Kny M, Böl G-F. Redox events in interleukin-1 signaling. Arch Biochem Biophys. 2004;423(1):66–73. doi: 10.1016/j.abb.2003.12.008 [DOI] [PubMed] [Google Scholar]
  • 136.Nagaya T, Fujieda M, Otsuka G, Yang J-P, Okamoto T, Seo H. A potential role of activated NF-κB in the pathogenesis of euthyroid sick syndrome. J Clin Invest. 2000;106(3):393–402. doi: 10.1172/JCI7771 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Tee LY, Harjanto S, Rosario BH. COVID-19 complicated by Hashimoto’s thyroiditis. Singapore Med J. 2020;62(5):265. doi: 10.11622/smedj.2020106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Ganesh BB, Bhattacharya P, Gopisetty A, Prabhakar BS. Role of cytokines in the pathogenesis and suppression of thyroid autoimmunity. J Interferon Cytokine Res. 2011;31(10):721–731. doi: 10.1089/jir.2011.0049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Pyzik A, Grywalska E, Matyjaszek-Matuszek B, Roliński J. Immune disorders in Hashimoto’s thyroiditis: what do we know so far? J Immunol Res. 2015;2015:1–8. doi: 10.1155/2015/979167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Mikoś H, Mikoś M, Obara-Moszyńska M, Niedziela M. The role of the immune system and cytokines involved in the pathogenesis of autoimmune thyroid disease (AITD). Endokrynol Pol. 2014;65(2):150–155. doi: 10.5603/EP.2014.0021 [DOI] [PubMed] [Google Scholar]
  • 141.Kochman J, Jakubczyk K, Bargiel P, Janda-Milczarek K. The influence of oxidative stress on thyroid diseases. Antioxidants. 2021;10(9):1442. doi: 10.3390/antiox10091442 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Corssmit E, De Metz J, Sauerwein H, Romijn J. Biologic responses to IFN-α administration in humans. J Interferon Cytokine Res. 2000;20(12):1039–1047. doi: 10.1089/107999000750053690 [DOI] [PubMed] [Google Scholar]
  • 143.Prummel MF, Wiersinga WM. Thyroid peroxidase autoantibodies in euthyroid subjects. Best Pract Res Clin Endocrinol Metab. 2005;19(1):1–15. doi: 10.1016/j.beem.2004.11.003 [DOI] [PubMed] [Google Scholar]
  • 144.Teillaud J-L. Antibody-dependent cellular cytotoxicity (ADCC). eLS; 2012.
  • 145.Wang SH, Baker JR Jr. The role of apoptosis in thyroid autoimmunity. Thyroid. 2007;17(10):975–979. doi: 10.1089/thy.2007.0208 [DOI] [PubMed] [Google Scholar]
  • 146.Tran SE, Holmström TH, Ahonen M, Kähäri V-M, Eriksson JE. MAPK/ERK overrides the apoptotic signaling from Fas, TNF, and TRAIL receptors. J Biol Chem. 2001;276(19):16484–16490. doi: 10.1074/jbc.M010384200 [DOI] [PubMed] [Google Scholar]
  • 147.Hussein MR, Haemel AK, Wood GS. Apoptosis and melanoma: molecular mechanisms. J Pathol. 2003;199(3):275–288. doi: 10.1002/path.1300 [DOI] [PubMed] [Google Scholar]
  • 148.Mezosi E, Wang SH, Utsugi S, et al. Induction and regulation of Fas-mediated apoptosis in human thyroid epithelial cells. Mol Endocrinol. 2005;19(3):804–811. doi: 10.1210/me.2004-0286 [DOI] [PubMed] [Google Scholar]
  • 149.W-p Z, Flavell RA. The transcription factor GATA-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells. Cell. 1997;89(4):587–596. doi: 10.1016/S0092-8674(00)80240-8 [DOI] [PubMed] [Google Scholar]
  • 150.Hiromatsu Y, Kaku H, Mukai T, et al. Immunohistochemical analysis of bcl-2, Bax and Bak expression in thyroid glands from patients with Graves’ disease. Endocr J. 2004;51(4):399–405. doi: 10.1507/endocrj.51.399 [DOI] [PubMed] [Google Scholar]
  • 151.Kuryłowicz A, Nauman J. The role of nuclear factor-kappaB in the development of autoimmune diseases: a link between genes and environment. Acta Biochim Pol. 2008;55(4):629–647. doi: 10.18388/abp.2008_3023 [DOI] [PubMed] [Google Scholar]
  • 152.Hirata Y, Takahashi M, Morishita T, Noguchi T, Matsuzawa A. Post-translational modifications of the TAK1-TAB complex. Int J Mol Sci. 2017;18(1):205. doi: 10.3390/ijms18010205 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Hanafusa T, Chiovato L, Doniach D, Pujol-Borrell R, Russell R, Bottazzo G. ABERRANT EXPRESSION OF HLA-DR ANTIGEN ON THYROCYTES IN GRAVES’DISEASE: RELEVANCE FOR AUTOIMMUNITY. Lancet. 1983;322(8359):1111–1115. doi: 10.1016/S0140-6736(83)90628-1 [DOI] [PubMed] [Google Scholar]
  • 154.Faure GC, Bensoussan-Lejzerowicz D, Bene MC, Aubert V, Leclere J. Coexpression of CD40 and class II antigen HLA-DR in Graves’ disease thyroid epithelial cells. Clin immunol immunopathol. 1997;84(2):212–215. doi: 10.1006/clin.1997.4391 [DOI] [PubMed] [Google Scholar]
  • 155.Zazzeroni F, Papa S, Algeciras-Schimnich A, et al. Gadd45β mediates the protective effects of CD40 costimulation against Fas-induced apoptosis. Blood. 2003;102(9):3270–3279. doi: 10.1182/blood-2003-03-0689 [DOI] [PubMed] [Google Scholar]
  • 156.Kolls JK, Lindén A. Interleukin-17 family members and inflammation. Immunity. 2004;21(4):467–476. doi: 10.1016/j.immuni.2004.08.018 [DOI] [PubMed] [Google Scholar]
  • 157.Ghoreschi K, Laurence A, Yang X-P, et al. Generation of pathogenic TH17 cells in the absence of TGF-β signalling. Nature. 2010;467:7318):967–971. doi: 10.1038/nature09447 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Lui DTW, Lee CH, Chow WS, et al. Thyroid dysfunction in relation to immune profile, disease status, and outcome in 191 patients with COVID-19. J Clin Endocrinol Metab. 2021;106(2):e926–e935. doi: 10.1210/clinem/dgaa813 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Huber AK, Finkelman FD, Li CW, et al. Genetically driven target tissue overexpression of CD40: a novel mechanism in autoimmune disease. J Immunol. 2012;189(6):3043–3053. doi: 10.4049/jimmunol.1200311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Woeller CF, Roztocil E, Hammond C, Feldon SE. TSHR signaling stimulates proliferation through PI3K/Akt and induction of miR-146a and miR-155 in thyroid eye disease orbital fibroblasts. Invest Ophthalmol Vis Sci. 2019;60(13):4336–4345. doi: 10.1167/iovs.19-27865 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Michalek K, Morshed SA, Latif R, Davies TF. TSH receptor autoantibodies. Autoimmun Rev. 2009;9(2):113–116. doi: 10.1016/j.autrev.2009.03.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Morshed SA, Latif R, Davies TF. Characterization of thyrotropin receptor antibody-induced signaling cascades. Endocrinology. 2009;150(1):519–529. doi: 10.1210/en.2008-0878 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Muller I, Cannavaro D, Dazzi D, et al. SARS-CoV-2-related atypical thyroiditis. Lancet Diabetes Endocrinol. 2020;8(9):739–741. doi: 10.1016/S2213-8587(20)30266-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Carella C, Mazziotti G, Amato G, Braverman L, Roti E. Interferon-α-related thyroid disease: pathophysiological, epidemiological, and clinical aspects. J Clin Endocrinol Metab. 2004;89(8):3656–3661. doi: 10.1210/jc.2004-0627 [DOI] [PubMed] [Google Scholar]
  • 165.Pearce EN, Farwell AP, Braverman LE. Thyroiditis. N Engl J Med. 2003;348(26):2646–2655. doi: 10.1056/NEJMra021194 [DOI] [PubMed] [Google Scholar]
  • 166.Truran P, Aspinall S. Thyrotoxicosis and thyroiditis. Surgery. 2014;32(10):537–542. doi: 10.1016/j.mpsur.2014.07.006 [DOI] [Google Scholar]
  • 167.Asfuroglu Kalkan E, Ates I. A case of subacute thyroiditis associated with Covid-19 infection. J Endocrinol Invest. 2020;43(8):1173–1174. doi: 10.1007/s40618-020-01316-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Volpé R. Thyroiditis: current views of pathogenesis. Med Clin North Am. 1975;59(5):1163–1175. doi: 10.1016/S0025-7125(16)31965-4 [DOI] [PubMed] [Google Scholar]
  • 169.Chakravarthy V, Ejaz S. Thyroxine-binding globulin deficiency; 2019. [PubMed]
  • 170.Di Marco F, Foti G, Corsico A. Where are we with the use of N-acetylcysteine as a preventive and adjuvant treatment for COVID-19. Eur Rev Med Pharmacol Sci. 2022;26(2):715–721. doi: 10.26355/eurrev_202201_27898 [DOI] [PubMed] [Google Scholar]
  • 171.Maia AL, Goemann IM, Meyer EL, Wajner SM. Deiodinases: the balance of thyroid hormone: type 1 iodothyronine deiodinase in human physiology and disease. J Endocrinol. 2011;209(3):283–297. doi: 10.1530/JOE-10-0481 [DOI] [PubMed] [Google Scholar]
  • 172.Lehnen TE, Santos MV, Lima A, Maia AL, Wajner SM. N-acetylcysteine prevents low T3 syndrome and attenuates cardiac dysfunction in a male rat model of myocardial infarction. Endocrinology. 2017;158(5):1502–1510. doi: 10.1210/en.2016-1586 [DOI] [PubMed] [Google Scholar]
  • 173.Marrocco I, Altieri F, Peluso I. Measurement and clinical significance of biomarkers of oxidative stress in humans. Oxid Med Cell Longev. 2017;2017. doi: 10.1155/2017/6501046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Vidart J, Wajner SM, Leite RS, et al. N-acetylcysteine administration prevents nonthyroidal illness syndrome in patients with acute myocardial infarction: a randomized clinical trial. J Clin Endocrinol Metab. 2014;99(12):4537–4545. doi: 10.1210/jc.2014-2192 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.De Flora S, Balansky R, La Maestra S. Rationale for the use of N‐acetylcysteine in both prevention and adjuvant therapy of COVID‐19. THE FASEB Journal. 2020;34(10):13185–13193. doi: 10.1096/fj.202001807 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Bhattacharya R, Mondal M, Naiya SB, Lyngdoh L, Mukherjee R, Singh PK. The beneficial role of N-acetylcysteine as an adjunctive drug in treatment of COVID-19 patients in a tertiary care hospital in India: an observational study. Int J Res Med Sci. 2020;8(10):1. doi: 10.18203/2320-6012.ijrms20204010 [DOI] [Google Scholar]
  • 177.Gianfrancesco M, Hyrich KL, Al-Adely S, et al. Characteristics associated with hospitalisation for COVID-19 in people with rheumatic disease: data from the COVID-19 Global Rheumatology Alliance physician-reported registry. Ann Rheum Dis. 2020;79(7):859–866. doi: 10.1136/annrheumdis-2020-217871 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Ungaro RC, Brenner EJ, Agrawal M, et al. Impact of medications on COVID-19 outcomes in inflammatory bowel disease: analysis of more than 6000 patients from an international registry. Gastroenterology. 2022;162(1):316–319.e315. doi: 10.1053/j.gastro.2021.09.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Leone GM, Mangano K, Petralia MC, Nicoletti F, Past FP. Present and (Foreseeable) Future of Biological Anti-TNF Alpha Therapy. J Clin Med. 2023;12(4):1630. doi: 10.3390/jcm12041630 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Queiroz NS, Martins CA, Quaresma AB, et al. COVID‐19 outcomes in patients with inflammatory bowel diseases in Latin America: results from SECURE‐IBD registry. J Gastroenterol Hepatol. 2021;36(11):3033–3040. doi: 10.1111/jgh.15588 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Sparks JA, Wallace ZS, Seet AM, et al. Associations of baseline use of biologic or targeted synthetic DMARDs with COVID-19 severity in rheumatoid arthritis: results from the COVID-19 Global Rheumatology Alliance physician registry. Ann Rheum Dis. 2021;80(9):1137–1146. doi: 10.1136/annrheumdis-2021-220418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Lin S, Lau LH, Chanchlani N, Kennedy NA, Ng SC. Recent advances in clinical practice: management of inflammatory bowel disease during the COVID-19 pandemic. Gut. 2022;71(7):1426–1439. doi: 10.1136/gutjnl-2021-326784 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Edelman-Klapper H, Zittan E, Shitrit AB-G, et al. Lower serologic response to COVID-19 mRNA vaccine in patients with inflammatory bowel diseases treated with anti-TNFα. Gastroenterology. 2022;162(2):454–467. doi: 10.1053/j.gastro.2021.10.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Scavone C, Mascolo A, Rafaniello C, et al. Therapeutic strategies to fight COVID‐19: which is the status artis? Br J Pharmacol. 2022;179(10):2128–2148. doi: 10.1111/bph.15452 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Bessler H, Mendel C, Straussberg R, Gurary N, Aloni D, Sirota L. Effects of dexamethasone on IL-1β, IL-6, and TNF-α production by mononuclear cells of newborns and adults. Neonatology. 1999;75(4):225–233. doi: 10.1159/000014099 [DOI] [PubMed] [Google Scholar]
  • 186.Oeckinghaus A, Ghosh S. The NF-κB family of transcription factors and its regulation. Cold Spring Harb Perspect Biol. 2009;1(4):a000034. doi: 10.1101/cshperspect.a000034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Aghai ZH, Kumar S, Farhath S, et al. Dexamethasone suppresses expression of Nuclear Factor-kappaB in the cells of tracheobronchial lavage fluid in premature neonates with respiratory distress. Pediatr Res. 2006;59(6):811–815. doi: 10.1203/01.pdr.0000219120.92049.b3 [DOI] [PubMed] [Google Scholar]
  • 188.Nelson G, Wilde GJ, Spiller DG, et al. NF-κB signalling is inhibited by glucocorticoid receptor and STAT6 via distinct mechanisms. J Cell Sci. 2003;116(12):2495–2503. doi: 10.1242/jcs.00461 [DOI] [PubMed] [Google Scholar]
  • 189.Group TRC. Dexamethasone in hospitalized patients with Covid-19—preliminary report. New Engl J Med. 2020;2020:1. [Google Scholar]
  • 190.Haugen BR. Drugs that suppress TSH or cause central hypothyroidism. Best Pract Res Clin Endocrinol Metab. 2009;23(6):793–800. doi: 10.1016/j.beem.2009.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Alkemade A, Unmehopa U, Wiersinga W, Swaab D, Fliers E. Glucocorticoids decrease thyrotropin-releasing hormone messenger ribonucleic acid expression in the paraventricular nucleus of the human hypothalamus. J Clin Endocrinol Metab. 2005;90(1):323–327. doi: 10.1210/jc.2004-1430 [DOI] [PubMed] [Google Scholar]
  • 192.Wilber JF, Utiger RD. The effect of glucocorticoids on thyrotropin secretion. J Clin Invest. 1969;48(11):2096–2103. doi: 10.1172/JCI106176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.John C, Christian H, Morris J, Flower R, Solito E, Buckingham J. Kinase‐dependent regulation of the secretion of thyrotrophin and luteinizing hormone by glucocorticoids and annexin 1 peptides. J Neuroendocrinol. 2003;15(10):946–957. doi: 10.1046/j.1365-2826.2003.01081.x [DOI] [PubMed] [Google Scholar]
  • 194.Cote-Vélez A, Pérez-Martínez L, Charli J-L, Joseph-Bravo P. The PKC and ERK/MAPK pathways regulate glucocorticoid action on TRH transcription. Neurochem Res. 2008;33(8):1582–1591. doi: 10.1007/s11064-008-9698-5 [DOI] [PubMed] [Google Scholar]
  • 195.Zhao L, Su J, Liu S, et al. MAP kinase phosphatase MKP-1 regulates p-ERK1/2 signaling pathway with fluoride treatment. Biochem Biophys Res Commun. 2021;542:65–72. doi: 10.1016/j.bbrc.2020.12.100 [DOI] [PubMed] [Google Scholar]
  • 196.Kassel O, Sancono A, Krätzschmar J, Kreft B, Stassen M, Cato AC. Glucocorticoids inhibit MAP kinase via increased expression and decreased degradation of MKP-1. EMBO J. 2001;20(24):7108–7116. doi: 10.1093/emboj/20.24.7108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Joseph-Bravo P, Jaimes-Hoy L, Charli J-L. Regulation of TRH neurons and energy homeostasis-related signals under stress. J Endocrinol. 2015;224(3):R139–R159. doi: 10.1530/JOE-14-0593 [DOI] [PubMed] [Google Scholar]
  • 198.Luo W, Li Y-X, Jiang L-J, Chen Q, Wang T, Ye D-W. Targeting JAK-STAT signaling to control cytokine release syndrome in COVID-19. Trends Pharmacol Sci. 2020;41(8):531–543. doi: 10.1016/j.tips.2020.06.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Satarker S, Tom AA, Shaji RA, Alosious A, Luvis M, Nampoothiri M. JAK-STAT pathway inhibition and their implications in COVID-19 therapy. Postgrad Med. 2021;133(5):489–507. doi: 10.1080/00325481.2020.1855921 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Keystone EC, Taylor PC, Drescher E, et al. Safety and efficacy of baricitinib at 24 weeks in patients with rheumatoid arthritis who have had an inadequate response to methotrexate. Ann Rheum Dis. 2015;74(2):333–340. doi: 10.1136/annrheumdis-2014-206478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Sorrell FJ, Szklarz M, Azeez KRA, Elkins JM, Knapp S. Family-wide structural analysis of human numb-associated protein kinases. Structure. 2016;24(3):401–411. doi: 10.1016/j.str.2015.12.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Richardson PJ, Corbellino M, Stebbing J. Baricitinib for COVID-19: a suitable treatment?–Authors’ reply. Lancet Infect Dis. 2020;20(9):1013–1014. doi: 10.1016/S1473-3099(20)30270-X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Stebbing J, Phelan A, Griffin I, et al. COVID-19: combining antiviral and anti-inflammatory treatments. Lancet Infect Dis. 2020;20(4):400–402. doi: 10.1016/S1473-3099(20)30132-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Gillenwater S, Rahaghi F, Hadeh A. Remdesivir for the treatment of Covid-19-preliminary report. N Engl J Med. 2020;383(10):992. [DOI] [PubMed] [Google Scholar]
  • 205.Mullard A. Ebola outbreak prompts experimental drug rollout. Nat Rev Drug Discov. 2018;17(7):460–461. [DOI] [PubMed] [Google Scholar]
  • 206.Agostini ML, Andres EL, Sims AC, et al. Coronavirus susceptibility to the antiviral remdesivir (GS-5734) is mediated by the viral polymerase and the proofreading exoribonuclease. MBio. 2018;9(2):e00221–00218. doi: 10.1128/mBio.00221-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Warren TK, Jordan R, Lo MK, et al. Therapeutic efficacy of the small molecule GS-5734 against Ebola virus in rhesus monkeys. Nature. 2016;531(7594):381–385. doi: 10.1038/nature17180 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Arshad U, Pertinez H, Box H, et al. Prioritization of anti‐SARS‐Cov‐2 drug repurposing opportunities based on plasma and target site concentrations derived from their established human pharmacokinetics. Clin Pharmacol Ther. 2020;108(4):775–790. doi: 10.1002/cpt.1909 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Kalil AC, Patterson TF, Mehta AK, et al. Baricitinib plus remdesivir for hospitalized adults with Covid-19. N Engl J Med. 2021;384(9):795–807. doi: 10.1056/NEJMoa2031994 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Vitiello A, Ferrara F. Perspectives of association Baricitinib/Remdesivir for adults with Covid-19 infection. Mol Biol Rep. 2022;49(1):827–831. doi: 10.1007/s11033-021-06888-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Kermali M, Khalsa RK, Pillai K, Ismail Z, Harky A. The role of biomarkers in diagnosis of COVID-19–A systematic review. Life Sci. 2020;254:117788. doi: 10.1016/j.lfs.2020.117788 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Poggiali E, Zaino D, Immovilli P, et al. Lactate dehydrogenase and C-reactive protein as predictors of respiratory failure in CoVID-19 patients. Clin Chim Acta. 2020;509:135–138. doi: 10.1016/j.cca.2020.06.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Sabbatino F, Conti V, Franci G, et al. PD-L1 dysregulation in COVID-19 patients. Front Immunol. 2021;12:695242. doi: 10.3389/fimmu.2021.695242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Sambugaro G, Brambilla E, Costanzo G, et al. COVID-19 Clinical Features and Outcome in Italian Patients Treated with Biological Drugs Targeting Type 2 Inflammation. Life. 2024;14(3):378. doi: 10.3390/life14030378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Penman SL, Kiy RT, Jensen RL, et al. Safety perspectives on presently considered drugs for the treatment of COVID‐19. Br J Pharmacol. 2020;177(19):4353–4374. doi: 10.1111/bph.15204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Marinella MA. Routine antiemetic prophylaxis with dexamethasone during COVID-19: should oncologists reconsider? J Oncol Pharm Pract. 2020;26(6):1482–1485. doi: 10.1177/1078155220931921 [DOI] [PubMed] [Google Scholar]
  • 217.Prentice RE, Al‐Ani A, Christensen B. Managing COVID‐19 in patients with inflammatory bowel disease: navigating unprecedented challenges. Intern Med J. 2021;51(2):284–287. doi: 10.1111/imj.15190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Y-N N, Chen G, Sun J, Liang B-M, Liang Z-A. The effect of corticosteroids on mortality of patients with influenza pneumonia: a systematic review and meta-analysis. Critical Care. 2019;23(1):1–9. doi: 10.1186/s13054-018-2293-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Dixit D, Kuete NT, Bene P, Khan I, Oprea-Ilies G, Flenaugh E. invasive pulmonary aspergillosis with hydropneumothorax in a patient taking high-dose glucocorticoids. Am J Case Rep. 2020;21:e928499–928491. doi: 10.12659/AJCR.928499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Olivera MJ. Dexamethasone and COVID-19: strategies in low-and middle-income countries to tackle steroid-related Strongyloides hyperinfection. American JTrop Med Hyg. 2021;104(5):1611. doi: 10.4269/ajtmh.20-1085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Chiu MN, Bhardwaj M, Sah SP. Safety profile of COVID-19 drugs in a real clinical setting. Eur J Clin Pharmacol. 2022;2022:1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Gupte V, Hegde R, Sawant S, et al. Safety and clinical outcomes of remdesivir in hospitalised COVID-19 patients: a retrospective analysis of active surveillance database. BMC Infect Dis. 2022;22(1):1–8. doi: 10.1186/s12879-021-07004-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.W-j G, Z-y N, Hu Y, et al. Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med. 2020;382(18):1708–1720. doi: 10.1056/NEJMoa2002032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Bangash MN, Patel J, Parekh D. COVID-19 and the liver: little cause for concern. Lancet Gastroenterol Hepatol. 2020;5(6):529–530. doi: 10.1016/S2468-1253(20)30084-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Barkas F, Styla C-P, Bechlioulis A, Milionis H, Liberopoulos E. Sinus bradycardia associated with remdesivir treatment in COVID-19: a case report and literature review. J Cardiovasc Develop Dis. 2021;8(2):18. doi: 10.3390/jcdd8020018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Peng L, Xiao K, Ottaviani S, Stebbing J, Wang Y-J. A real-world disproportionality analysis of FDA Adverse Event Reporting System (FAERS) events for baricitinib. Expert Opin Drug Saf. 2020;19(11):1505–1511. doi: 10.1080/14740338.2020.1799975 [DOI] [PubMed] [Google Scholar]
  • 227.Smolen JS, Genovese MC, Takeuchi T, et al. Safety profile of baricitinib in patients with active rheumatoid arthritis with over 2 years median time in treatment. J Rheumatol. 2019;46(1):7–18. doi: 10.3899/jrheum.171361 [DOI] [PubMed] [Google Scholar]
  • 228.Taylor PC, Weinblatt ME, Burmester GR, et al. Cardiovascular safety during treatment with baricitinib in rheumatoid arthritis. Arthritis Rheumatol. 2019;71(7):1042–1055. doi: 10.1002/art.40841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Klok F, Kruip M, Van der Meer N, et al. Confirmation of the high cumulative incidence of thrombotic complications in critically ill ICU patients with COVID-19: an updated analysis. Thromb Res. 2020;191:148–150. doi: 10.1016/j.thromres.2020.04.041 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Drug Design, Development and Therapy are provided here courtesy of Dove Press

RESOURCES