Next Article in Journal
Mesocricetus auratus (Golden Syrian Hamster) Experimental Model of SARS-CoV-2 Infection Reveals That Lung Injury Is Associated with Phenotypic Differences Between SARS-CoV-2 Variants
Previous Article in Journal
Hemophagocytic Lymphohistiocytosis Triggered by Dengue: A Narrative Review and Individual Patient Data Meta-Analysis
Previous Article in Special Issue
Beyond Stress Granules: G3BP1 and G3BP2 Redundantly Suppress SARS-CoV-2 Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of SARS-CoV-2 Nucleocapsid Protein in Host Inflammation

1
Department of Food Science and Technology, National University of Singapore, Singapore 117542, Singapore
2
Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
3
Biomedical and Health Technology Platform, National University of Singapore (Suzhou) Research Institute, Suzhou 215123, China
*
Author to whom correspondence should be addressed.
Viruses 2025, 17(8), 1046; https://doi.org/10.3390/v17081046
Submission received: 26 June 2025 / Revised: 22 July 2025 / Accepted: 25 July 2025 / Published: 27 July 2025
(This article belongs to the Special Issue Viral Mechanisms of Immune Evasion)

Abstract

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has posed substantial health threats and triggered widespread global economic disruption. The nucleocapsid (N) protein of SARS-CoV-2 is not only a key structural protein but also instrumental in mediating the host immune response, contributing significantly to inflammation and viral pathogenesis. Due to its immunogenic properties, SARS-CoV-2 N protein also interacts with host factors associated with various pre-existing inflammatory conditions and may possibly contribute to the long-term symptoms suffered by some COVID-19 patients after recovery—known as long COVID. This review provides a comprehensive overview of recent advances in elucidating the biological functions of the N protein. In particular, it highlights the mechanisms by which the N protein contributes to host inflammatory responses and elaborates on its association with long COVID and pre-existing inflammatory disorders.

1. Introduction

COVID-19 is caused by SARS-CoV-2, which is a positive-sense, single-stranded RNA virus. Millions of COVID-19 cases have been recorded globally, posing significant challenges for public health and inflicting considerable economic damage [1]. Similar to SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV), which are responsible for two drastic outbreaks, SARS-CoV-2 is a member of the Coronaviridae family [2]. Based on their genetic and serological features, coronaviruses (CoVs) are divided into four genera, namely α-, β-, γ-, and δ-coronaviruses [3]. Among these, α- and β-CoVs mainly infect mammals and pose serious threats to humans. SARS-CoV, MERS-CoV, SARS-CoV-2, human coronavirus (HCoV)-OC43, and HCoV-HKU1 are classified as β-CoVs; HCoV-229E and HCoV-NL63, on the other hand, are members of the alpha-CoVs [3].
The SARS-CoV-2 virion is primarily composed of the spike (S) protein, envelope (E) protein, membrane (M) protein, N protein, and viral RNA. Its genome contains 14 functional open reading frames (ORFs), with ORF1a and ORF1b occupying the majority of the genome and encoding 16 nonstructural proteins (NSPs) that are essential for viral replication. Additionally, nine accessory proteins contribute to host immune evasion and promote viral replication [4]. Importantly, the four structural proteins play significant functions in viral entry, assembly, and replication. Among them, the N protein plays key roles in viral mRNA transcription and replication, in addition to being involved in modulating immune responses and viral pathogenesis [5]. Therefore, it has been developed as a potential target for therapeutic development and diagnostic applications in COVID-19.
This review highlights recent findings on the modulatory functions of SARS-CoV-2 N protein across multiple stages of the viral life cycle, as well as its critical role in regulating host immune responses. In addition, the possible contribution of the N protein to long COVID and the interplay between the N protein and other pre-existing inflammatory diseases are also summarized.

2. N Protein

2.1. N Protein Structure

The SARS-CoV-2 N protein is a multidomain RNA-binding protein comprising 419 amino acids [6]. Two conserved and functional structural domains, the N-terminal domain (NTD) and C-terminal domain (CTD), are linked by intrinsically disordered regions (IDRs), including the central linking region (LKR), which contains a Ser/Arg (SR)-rich region that serves as a putative phosphorylation site. In addition, the N protein is flanked by two other LKRs, referred to as the N-arm and C-tail [6].

2.2. N Protein and Viral Life Cycle

SARS-CoV-2 entry into host cells is orchestrated by the S protein through endocytosis or direct fusion with the cell membrane [7]. After entry, the N protein is liberated into the cytoplasmic compartment, where viral genomic RNA dissociates from the N protein and undergoes replication and translation [8]. Newly synthesized viral genomes bound with N protein are assembled into enveloped particles with other structural proteins within the lumen of the endoplasmic reticulum–Golgi intermediate compartment (ERGIC) [8]. The progeny virions are then transported from the ERGIC to the Golgi apparatus and ultimately to the membrane, where they are released through exocytosis or lysosomal trafficking [8].
The SARS-CoV-2 N protein is essential for forming a helical ribonucleoprotein (RNP) complex with the RNA genome, mediated by regions with high positive charge on the surfaces of both NTD and CTD [9,10]. The RNP is crucial for viral replication and transcription, as it facilitates virion assembly and shields the virus from the dynamic immune response [9]. The interaction between the N protein and viral RNA, driven by liquid–liquid phase separation (LLPS), results in the formation of membraneless biomolecular condensates, which play key roles in multiple steps of the viral life cycle [11].
During LLPS, 5′-proximal genomic RNA transcripts of SARS-CoV-2 undergo conformational changes, mediated by the stem-loop (SL) SL1 and SL5a/b/c. These SL conformers are preferentially bound by the N protein [12]. Through LLPS, the N protein promotes the assembly of the RNA polymerase (RdRp) complex, including NSP7, NSP8, NSP12, and polyU RNA, ensuring high initiation and elongation rates during viral transcription [13]. A high-frequency trinucleotide polymorphism (GGG-to-AAC) variation leads to amino acid substitutions such as NR203K/G204R, significantly enhancing LLPS formation [14]. This mutation also promotes N protein phosphorylation and confers resistance to glycogen synthase kinase-3 (GSK-3) inhibition, thereby facilitating viral replication [15]. The N protein is recruited to the RTCs during genome replication by binding to the Ubl1 domain of NSP3, enhancing protein–RNA interactions either through enzymatic activities or by stabilizing the complex structure [16]. In early infection, phosphorylation of the SR-rich region of N protein by cytoplasmic kinase regulates the function of N protein. This modification facilitates RNA structural rearrangements required for the transcription of long subgenomic RNAs in the RTC by interacting with DEAD-box helicase 1 (DDX1) [17,18]. Additionally, it is discovered that the N-CTD plays a key part in interactions with the M protein during the budding process, as well as in forming the RNA-binding groove required for viral assembly [19]. The N protein also binds strongly to anionic lipids, such as phosphoinositide and phosphatidylserine on the membranes, via its CTD, which supports its localization to assembly sites [20]. The E3 ubiquitin ligase tripartite motif protein 6 (TRIM6) binds to N-CTD through its RING and B-box-CCD domains and modulates the K29-typed polyubiquitination of N-NTD (K102) and N-CTD (K347 and K361) [21]. The ubiquitination enhances the ability of N protein to bind to viral genomic RNA, thereby promoting viral propagation [21]. The N-C tail of N protein contributes to the encapsidation of genomic RNA, which may be regulated by interactions with the M protein [18]. Since viruses cannot produce ATP independently, they rely entirely on the host cell’s ATP as an energy source for key stages of their life cycle [22]. Intriguingly, the N-CTD has recently been identified as a novel ATP-binding site, suggesting its role in modulating phase separation [22].

3. N Protein and Inflammation

3.1. N Protein and Innate Immunity

3.1.1. N Protein and Intracellular Inflammatory Signaling

The innate and adaptive immune systems are two major types of immune responses in the integrated human immune system. Innate immunity is activated when pattern recognition receptors (PRRs) detect viral pathogen-associated molecular patterns (PAMPs). Among the PRRs, the retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), including RIG-I and melanoma differentiation-associated gene 5 (MDA5), belonging to RLRs necessarily work in sensing cytoplasmic dsRNA during infection [23,24]. Upon activation, these receptors undergo conformational rearrangement and oligomerization, which initiates mitochondrial antiviral signaling (MAVS), thereby triggering a cascade of immune responses, such as the NF-κB, IRF3 and IRF7 pathways. These cascades ultimately lead to the production of type I interferon (IFN-I) and pro-inflammatory cytokines like IL-6 and IFN-α/β [25,26]. The N protein dampens host defenses and facilitates immune evasion by suppressing and modulating multiple components of the innate immune response (Figure 1).
RIG-I is a key cytosolic pattern recognition receptor that detects viral RNA and initiates the activation of IFN-I and other antiviral genes, thereby orchestrating the host innate immune response. TRIM25-mediated ubiquitination of RIG-1 is inhibited by the association between SARS-CoV N-CTD and the SPRY domain of TRIM25 [27]. Likewise, N protein associates with TRIM25, resulting in the inhibition of TRIM25-dependent IFN production [28]. N protein also inhibits polyinosinic: polycytidylic acid [poly(I:C)]-facilitated IFN signaling by targeting tank-binding kinase 1 (TBK1), thereby preventing IRF3 undergoing nuclear translocation [28]. Additionally, it interacts with the RIG-I protein via the enzymatically active DExD/H domain, repressing the IFN-β response [29]. A study demonstrated that mutations at R203 and D377 enhance the N protein-mediated impairment of RIG-I signaling, including reduced IRF3 phosphorylation and IFN-β release [30].
Stress granules (SGs) are cytoplasmic, electron-dense, membraneless structures formed in response to viral infection as part of the host’s antiviral defense mechanism [31]. The N protein has been proven to suppress antiviral SG formation by interacting with GTPase-activating protein-binding protein 1 (G3BP1), which has a critical function in RIG-I recognition of pathogenic RNA [32,33,34]. During viral replication, the N protein undergoes LLPS, which suppresses SG assembly and inhibits IFN-I production [34]. Moreover, the interaction between G3BP1 and the N protein is enhanced by N protein self-deacetylation, mediated through the induction of histone deacetylase 6 (HDAC6), a component of cytoplasmic SGs [35]. The methionine codon located at position 210 of the SARS-CoV-2 N protein (N*M210) is crucial for dsRNA binding and in inhibiting multiple aspects of the cellular antiviral response, such as SGs formation [36]. Furthermore, by sequestering growth arrest and DNA damage-inducible 34 (GADD34) mRNA into the N+foci and hindering its association with G3BP1, the N protein impedes IFN gene transcription and compromises the host innate immune response [37]. The SARS-CoV-2 N protein is known to interact with several proteins involved in immune responses, including activating signal co-integrator 1 complex subunit 3 (ASCC3), inosine monophosphate dehydrogenase 2 (IMPDH2), and adaptor-related protein complex 3 subunit beta 1 (AP3B1) [35].
Post-translational modification plays a pivotal role in regulating the effects of the SARS-CoV-2 N protein on the innate antiviral immune response. Previous research has shown that SARS-CoV-2 N protein represses the antiviral immune response by inhibiting Lys63-linked polyubiquitination and aggregation of MAVS. Furthermore, acetylation and deacetylation of Lys375 in the N-CTD region, mediated by host acetyltransferases and deacetylases, have been shown to inhibit MAVS signaling [38]. The N-CTD also serves a vital function in mediating liquid phase interaction with MAVS and inhibiting the MAV-mediated IFN response [39,40]. Poly (dA:dT), a RIG-I ligand, can induce the conjugation of the small ubiquitin-like modifiers (SUMO) to MAVS, thereby enhancing MAVS clustering and promoting the release of IFN-β. Suppression of the SUMOylation via ubiquitin-conjugating enzyme 9 (UBC9) siRNA significantly reduces the generation of poly(dA:dT)-induced IFN-β [41]. The SARS-CoV-2 N protein modulates MAVS SUMOylation by enhancing the interaction between UBC9 and MAVS, thereby interrupting IFN-β synthesis through suppression of IκB kinase-α (IKKα), TBK1, and IRF3 phosphorylation [42]. In addition, suppression of the innate antiviral immune response by the N protein is attributable to the SUMO conjugation on N protein lysine 65 residue. SUMOylation of N protein, facilitated by SUMO E3 ligase TRIM28, promotes its robust homo-oligomerization, RNA association, and LLPS activity [43]. Another E3 ligase, TRIM 21, mediates the polyubiquitination and degradation of N protein, thereby contributing to the regulation of host innate immunity [44]. Collectively, these findings demonstrate that post-translational SUMOylation of the N protein is critical for SARS-CoV-2 virulence and immune evasion. Surface proteins, including S, E, and M proteins, are easily glycosylated, while the N protein is less glycosylated but prone to phosphorylation [45]. Only two potential glycosylation sites on the N protein—N48 and N270—have been experimentally confirmed [46]. Glycosylation at these sites may mask epitopes, potentially interfering with host inflammatory responses; however, further evidence is still needed to substantiate these effects.
Furthermore, the underlying molecular mechanisms by which the SARS-CoV-2 N protein regulates the innate antiviral immune response have been increasingly elucidated, with NF-κB signaling emerging as one of the most prominent pathways activated in infected cells. Silencing NF-κB transcription factor p65 or p50 disrupts NF-κB signaling and reduces viral replication, an effect that is reversed upon their re-expression [47]. Intriguingly, N protein facilitates NF-κB response and cytokine production. Studies have found that N protein strongly activates NF-κB signaling pathways through toll-like receptor 2 (TLR2) and the mitogen-activated protein kinase (MAPK) pathway in human endothelial cells [48]. In HEK293T cells, N protein overexpression leads to cytoplasmic changes and increased nuclear accumulation of NF-κB p65 [49]. In murine macrophages, N protein not only regulates the secretion of pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-10, but also alters the expression of immune-related genes in Janus kinase-signal transducer and activator of transcription (JAK-STAT), TNF, NF-κB, and MAPK signaling pathways [50]. A similar pattern of elevated cytokine expression is observed in N protein-stimulated lung epithelial A549 cells and the serum of COVID-19 patients [51]. The development of a cytokine storm, characterized by excessive production of cytokines such as IL-6 and poor type I IFN induction, is a hallmark of severe COVID-19 [52]. In murine models, N protein stimulation leads to acute lung injury marked by strong NF-κB activation and an exaggerated pro-inflammatory cytokine response [53]. Mechanistically, one reason for the interplay between N protein and NF-κB signaling may lie in the N protein’s ability to recruit key NF-κB pathway kinases, including TAK1 and IKK complex, during LLPS with RNA, thereby enhancing NF-κB activation [54]. In addition, the N protein binds to the receptor for advanced glycation end products (RAGE) via its NTD and CTD, activating the extracellular signal-regulated kinases 1 and 2 (ERK1/2)–NF-κB pathway through RAGE [55]. Interestingly, some genes show opposing expression patterns when NF-κB p65 is knocked down versus when the N protein is overexpressed, suggesting that N protein may trigger a distinct transcriptional program to modulate or fine-tune the NF-κB response [49].
Inflammasomes are multiprotein machinery that engage in innate antiviral immunity. They typically consist of adaptor apoptosis-associated speck-like protein-containing CARD (ASC), pro-caspase-1, and inflammasome nucleators such as nucleotide-binding oligomerization domain protein (NOD)-like receptors (NLRs), AIM2, and pyrin. Among the various identified inflammasomes, the NLR family pyrin domain containing-3 (NLRP3) inflammasome is the best understood because of its pivotal role in orchestrating inflammatory and antiviral mechanisms. Upon assembly, the NLRP3 inflammasome promotes pro-caspase-1 cleavage, which subsequently cleaves gasdermin D (GSDMD), inducing GSDMD-dependent pyroptosis. It also assists in the production of proinflammatory IL-18 and IL-1β [56]. The N protein has been shown to directly associate with NLRP3, enhancing its interaction with ASC and promoting NLRP3 inflammasome assembly. Remarkedly, lung damage and proinflammatory cytokine responses induced by the N protein can be mitigated by NLRP3 or caspase-1 inhibitors [57]. Another study demonstrated that N protein also directly interacts with the linker region of GSDMD, preventing its cleavage by caspase-1 and subsequently suppressing pyroptosis in SARS-CoV-2-infected human monocytes [58].
RNA interference (RNAi) plays critical antiviral defense roles in a wide range of organisms, including mammalian cells. It is a post-transcriptional gene silencing process initiated by double-stranded RNA (dsRNA), which is cleaved into small interfering RNA (siRNA) duplexes by the Dicer, an enzyme from the RNase III family of ATP-dependent ribonucleases [59,60]. At the effector phrase, these siRNA duplexes are incorporated into the RNA-induced silencing complex (RISC), guiding gene expression silencing [61]. The N protein suppresses the RNAi at both initiation and effector stages by obstructing siRNA biogenesis, RISC assembly, and target RNA cleavage. This enables the virus to evade host inflammatory responses and promotes viral replication in host cells [61]. In SARS-CoV, residue Lys 258 and Lys 262 with a positive charge in N-CTD were identified as critical residues for viral suppressors of RNAi (VSRs) [62]. A similar VSR region is presumed to exist in the N protein, likely within the CTD, though the exact residues remain unidentified. Another important antiviral mechanism is nonsense-mediated mRNA decay (NMD), which regulates and degrades aberrant mRNA to prevent the translation of viral transcripts. The N protein counteracts NMD by directly interacting with RNA helicase UP-frameshift-1 (UPF1) and UPF2, which are essential components in NMD. This interaction restrains the unwinding activity of UPF1 and disrupts the UPF1–UPF2 interaction, thereby inhibiting NMD and facilitating viral gene expression [63].
The cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP-dependent Cl channel that regulates the host immune defense against pathogen infection, such as SARS-CoV-2 [64]. The N protein downregulates CFTR expression by directly interacting with Smad3 via microRNA-145, leading to elevated intracellular Cl levels. In addition, the N protein further increases the intracellular Cl concentration by depleting intracellular cAMP by increasing the expression of phosphodiesterase 4 (PDE4). Consequently, the high concentration of intracellular Cl triggers serum glucocorticoid-regulated kinase 1 (SGK1) phosphorylation, subsequently activating a vigorous inflammatory response [65].
IFN-stimulated gene 15 (ISG15) is a ubiquitin-like protein that becomes covalently conjugated to host and viral proteins through intracellular enzymes such as HECT and RLD domain containing E3 ubiquitin protein ligase 5 (HERC5) [66]. The ISGylation is highly associated with the innate immune response against several viruses [67]. The N protein, particularly in its phosphorylated form, is subject to ISGylation by HERC5. This modification disrupts the N protein’s ability to form functional oligomers, ultimately inhibiting viral RNA synthesis [68]. Residues K261, K266, K355, K374, K387, and K388 within the N-CTD and spacer B/N3 domain are responsible for the ISGylation of N protein [67,68,69]. Interestingly, this antiviral ISGylation mechanism is counteracted by the SARS-CoV-2 papain-like protease (PLpro) through its deISGylating activity, thereby contributing to host antiviral immunity evasion [67,68].
The lectin pathway is a vital component of innate immunity, responsible for initiating the complement system, which is an immune surveillance mechanism that enables rapid detection and response to infections [70]. One study demonstrated that N protein aggravates inflammation by promoting overactivation of the lectin pathway through mannan-binding lectin serine protease 2 (MASP-2) overactivation [71]. However, other studies have shown that the N protein binds to neither MASP-1 nor MASP-2, nor does it activate the lectin pathway in normal human serum; instead, it undergoes proteolytic degradation when incubated with MASP-2 [72,73].
Although the N protein predominantly localizes in the cytoplasm, it is also detectable in the nucleus of infected cells, where it regulates gene expression and interferes with immune responses. Its nuclear translocation occurs via the nuclear pore complex and is mediated by interactions with Ras-related nuclear protein (RAN) and import receptors karyopherin alpha (KPNA) and karyopherin beta 1 (KPNB1) [66].
Importantly, the regulation of the innate immune response by the N protein is dose-dependent: a low concentration of the N protein inhibits IFN-I signaling and the secretion of inflammatory markers, while a high concentration of the N protein enhances the IFN-I pathway and the release of pro-inflammatory markers [74]. As SARS-CoV-2 evolves, subvariants with enhanced immune evasion capabilities tend to become dominant due to their ability to reduce innate immune activation. In response, the host undergoes convergent evolution to enhance innate immune antagonist expression, thereby counteracting viral immune escape [75].

3.1.2. N Protein and Systemic Inflammation

SARS-CoV-2 infection also triggers body-wide systemic inflammation, which can damage tissues and organs if not controlled. The influence of the N protein on systemic inflammation is illustrated in Figure 2. A drastic cytokine storm, characterized by considerably increased IL-6, IL-8, IL-10, TNF-α, and IFN levels in COVID-19 patients, leads to systemic inflammatory immune responses [76]. N protein induces the production of tumor necrosis factor receptor 2 (TNFR2), CCL4, IL-1β, IL-6, IL-6R, IL-10, and IL-12 in human monocytes [77]. Notably, N protein treatment results in the escalation of IL-6 mRNA expression, which might be attributable to the increased levels of NF-κB pathway markers like NF-κB subunit 1 (NFKB1) and NF-κB inhibitor alpha (NFKBIA) [77]. The LLPS of N protein also activates NF-κB and further facilitates robust secretion of IL-6, IL-1β, TNF-α, and CXCL10 [54]. While inflammasome activation does not directly drive the cytokine storm, it triggers the release of IL-1β, which, in turn, induces the production of IL-6 and IL-8 [78]. Mechanistically, pathogenic T helper type 1 (Th1) cells and CD14+CD16+ monocytes trigger proinflammatory signaling, leading to macrophage and neutrophil infiltration and a subsequent cytokine storm in the lungs [79]. IL-6 is a key cytokine in the COVID-19 cytokine storm and serves as an independent predictor of lung injury severity [80]. Its signaling has been linked to coagulopathy and hepatic endotheliopathy in COVID-19 pneumonia, contributing to the progression of liver injury [81].
Plasma levels of SARS-CoV-2 N antigen are associated with RAGE, IL-10, and IFN-gamma-inducible protein 10 kD (IP-10) levels, suggesting a role in modulating the progression to severe disease in hospitalized COVID-19 patients [82]. In a study involving 326 patients, 95% had plasma SARS-CoV-2 N antigen levels of at least 3 pg/mL, and 50% had levels equal to or exceeding 1000 pg/mL [83]. In another study, N protein concentrations in blood samples had a mean of 1734 pg/mL among 131 PCR-positive inpatients, compared to a mean of 157 pg/mL among 43 PCR-positive outpatients, indicating significantly higher N protein levels in hospitalized patients [84]. It has been demonstrated that the median plasma N antigen level is 7673 pg/mL in severe COVID-19 patients, 351.6 pg/mL in moderately affected patients, and 4.6 pg/mL in asymptomatic individuals [85]. The primary clinical outcome indicates that each 500 pg/mL increase in plasma N antigen level is associated with elevated inflammatory markers [82]. The exact mechanism by which the N protein exits the cell remains unclear; however, one plausible explanation is programmed inflammatory cell death, which may facilitate the release of intracellular contents, including unassembled viral proteins, through cell membrane rupture [86]. Extracellular vesicles, produced by most living cells, have the ability to transport viral proteins within a spherical lipid bilayer and are commonly found in blood, plasma, and serum, suggesting a possible mechanism for the dissemination of the N protein to the extracellular environment [87].
Clinically, COVID-19 patients admitted to the intensive care unit (ICU) exhibit elevated serum SARS-CoV-2 N antigen levels, which are associated with tissue damage biomarkers and heightened lung inflammatory responses [88,89]. C-reactive protein (CRP), produced in response to inflammation, activates the complement system and enhances phagocytosis, promoting the clearance of pathogens [90]. Elevated serum CRP levels in COVID-19 patients are positively associated with both circulating N antigen levels and COVID-19 severity [88,91]. Additionally, tissue damage biomarkers such as serum amyloid A (SAA) and ferritin are significantly elevated in COVID-19 patients with detectable N antigen, whereas procalcitonin (PCT) levels remain comparable to those of antigenemia-negative individuals [89]. The human humoral fluid-phase pattern recognition molecule (PRM) long pentraxin 3 (PTX3), a key anti-inflammatory component in innate immunity, has been shown to bind to the NTD of N proteins [92]. Although PTX3 does not effectively interfere with the viral life cycle, its serum levels show significant correlations with clinical inflammatory markers such as IL-6, IL-8, IL-10, CRP, total leukocyte count, neutrophil-to-lymphocyte ratio, and ferritin [92,93]. These associations support the idea that PTX3 binding to the N protein may contribute to disease severity in COVID-19 patients.
In addition to the lungs, the N protein has also been detected in various other organs, including the kidneys, heart, liver, colon, and small intestine, where it potentially contributes to both local and systemic inflammation (Figure 2) [94]. Remarkably, N protein in the hippocampus has been found to induce Tau phosphorylation, leading to cognitive impairment and associations with Alzheimer’s disease pathology [95]. This is attributed to the recruitment of Tau into SGs by N protein. However, the expression of SUMO2 can positively ameliorate N protein-induced Tau phosphorylation by directly promoting the SUMOylation of Tau or indirectly regulating the target kinase activity [95].
Although NSPs and accessory proteins are involved in modulating host inflammation, their primary role is immune evasion rather than the activation of inflammatory responses or direct association with sysmetic inflammation [4,96]. In contrast to the membrane-bound spike protein and the envelope protein embedded in the viral particle, the N protein is cytoplasmic and undergoes minimal glycosylation, making it more accessible to intracellular space and facilitating broad innate immune activation that contributes to systemic inflammation [45]. Therefore, the N protein plays a distinct pro-inflammatory role in host immune responses.

3.2. N Protein and Adaptive Immunity

Antibody responses to N protein have been shown to correlate with various demographic and clinical factors, such as anti-S antibody levels, smoking status, education level, income level, race/ethnicity, and age [97]. B-cell responses targeting the N protein can persist for up to six months following infection with COVID-19 [98]. Immunoglobulin M (IgM) specific to N protein peaks at around day nine after symptom onset, followed by a class switch to immunoglobulin G (IgG) [99]. Serum levels of anti-N antibodies remain detectable in convalescent individuals for up to two months post infection, although they decline significantly in most individuals after three months [100]. Notably, the serum level of IgG specific to N protein in recovered patients is positively associated with age [101]. Additionally, SARS-CoV-2 infection elicits durable cellular responses. Polyfunctional memory T cells remain detectable up to 12 months post infection [102]. During the onset of infection, the frequencies of circulating N-specific CD8+ and CD4+ T cells inversely correlate with upper airway viral loads and systemic inflammatory markers, suggesting that these N-specific T cell responses may be attributable to viral clearance prior to seroconversion [103]. CD8+ T cell and CD4+ T cell responses following natural infection have been reported to be stable for over 8 months [104]. CD8+ T cells targeting N protein are not only immunodominant but also more persistent than CD8+ T cells targeting S, E, and the matrix [105]. Notably, N-specific T cells have demonstrated protective roles in SARS-CoV-2 infection [106]. Recovered COVID-19 individuals carrying the human leukocyte antigen (HLA)-B7+ exhibit dominant CD8+ T cell responses targeting an epitope located in N-NTD [107]. High naive precursor frequency and promiscuous T cell receptor (TCR) αβ pairing in B7/N105+ CD8+ T cells support CD8+ T cell responses to immunodominant SARS-CoV-2 N epitopes. Moreover, CD8+ T cells recognizing N epitopes can be cross-reactive with circulating HCoV-OC43 and HCoV-HKU1 [107,108].

3.3. N Protein and Long COVID

Long COVID, or post-acute sequelae of SARS-CoV-2 infection (PASC), refers to a broad spectrum of new symptoms continuing for weeks or months after SARS-CoV-2 infection. The sequelae include cardiac arrhythmias, chronic respiratory failure, neuropsychiatric symptoms, and gastrointestinal symptoms [109,110]. Long COVID may be linked with ineffective clearance and prolonged immune activation by the adaptive immune system. Absence of increased immunity against the N protein, combined with its potent pro-inflammatory effect to stimulate cytokine and chemokine pathways, presumably contributes to the pathogenesis of long COVID (Figure 3) [111].
The N protein acts as a PAMP that activates multiple innate inflammatory signaling pathways (Figure 1), playing a significant role in triggering the cytokine storm, which is increasingly implicated in the development of long COVID. In patients with long COVID, a significantly increased number of N protein-specific CD4+ and CD8+ T cells that produce TNF-α and IL-2 has been observed [112]. It has been reported that cytokine storms characterized by elevated plasma IL-1β, IL-6, and TNF cytokine levels are associated with acute sequelae of COVID-19 [113]. Clinical inflammatory markers linked with N protein levels, such as elevated CRP and sustained levels of cytokines like IL-6, have also been associated with prolonged recovery time in COVID-19 patients [114]. Notably, findings from a systematic review and meta-analysis indicate the increased IL-6 levels alone are correlated with long COVID-19 [115]. Additionally, lower levels of initial anti-N IgG are associated with longer COVID symptom duration [110]. Antibodies specific to N protein and anti-N IgA concentrations have been detected in PASC patients who display higher humoral response during the acute stage, along with elevated levels of CD8+ T cells [116,117]. Moreover, individuals with PASC tend to experience a faster reduction in the prevalence of N-specific IFN-producing CD8+ T cells [104]. Specific immunological properties, marked by heightened CD4+ T cells and declined CD8+ memory T cells activation against the N-CTD, have been observed in neurologic PASC patients [118,119]. Therefore, N protein plays an active immunopathological role, spanning from acute infection to chronic post-acute sequelae.
The prolonged existence of N protein was detected in patients who succumbed to viral pneumonia, even 50 days after infection onset, indicating that the N protein does not degrade quickly but persists long after infection (Figure 3) [120]. Apart from the respiratory and immune system, N protein also impacts other tissues and disrupts hormonal balance at the post-infectious stage. N protein was identified in the conjunctiva, trabecular meshwork, and iris cells of a COVID-19 patient who was later diagnosed with acute angle-closure glaucoma and cataract after recovery [121]. The pathogenic mechanism of dacryoadenitis is potentially linked to SARS-CoV-2 as well, as inflammatory cells surrounding the lacrimal gland in a COVID-19 patient showed immunoreactivity to the N protein [122]. Anosmia, a long-lasting COVID-19 symptom accompanied by signs of local inflammation in the human olfactory epithelium, is putatively attributed to the presence of the N protein [123,124]. The possible mechanism is that interference with LLPS by the N protein, which suppresses the innate immune responses, contributes to long COVID [125]. The N protein has also been found in the gastrointestinal tract, and its presence is associated with morphological alterations of microvilli in the small intestinal wall, leading to bleeding ulceration in a patient who had recovered from COVID-19 [126]. Failure to clear N proteins may result in persistent immune responses with ongoing immune dysregulation, promoting the progression from acute infection to chronic sequelae.

3.4. N Protein and Pre-Existing Inflammatory Diseases

As SARS-CoV-2 induces severe inflammation in the host after infection, patients with pre-existing inflammatory disorders are at a higher risk of increased rates of hospitalizations and mortality [127,128]. Compared with healthy individuals, patients diagnosed with diabetes (type 1 and 2) or obesity do not have increased susceptibility to SARS-CoV-2 infection but do show elevated hospitalization rates and greater disease severity [129]. N protein has been identified in various organs (Figure 2), where it has the potential to exacerbate or interfere with existing inflammatory diseases [94]. The serum anti-N IgG titer of pneumonia patients was higher than that of the asymptomatic group after 6 months [101]. Therefore, the immunodominant function of the N protein may drive prolonged inflammation and contribute to poor outcomes in patients with inflammatory conditions such as diabetes. Patients with type 2 diabetes (T2D) display abnormally high levels of metabolites such as trimethylamine N-oxide (TMAO), which are associated with gut dysbiosis and can contribute to chronic inflammatory and degenerative diseases [130,131]. Interestingly, N proteins show synergistic effects with TMAO on NLRP3 inflammasome activation. This activation is regulated by the sterol regulatory element-binding proteins (SREBP) cleavage–activating protein (SCAP) pathway and associated with intracellular cholesterol homeostasis [132]. Hyperphosphatemia, a common complication of chronic kidney disease, drives the gathering of inorganic phosphate in the blood, which amplifies N protein-induced inflammation through the SCAP-SREBP pathway, facilitating the activation of the NLRP3 inflammasome [133]. The N protein also undergoes LLPS, during which an RNA-binding protein called endogenous transactive response-binding protein 43 kDa (TDP-43) is incorporated into the biomolecular condensate [134]. Since the presence of pathological glial cytoplasmic inclusions precedes fatal neurodegenerative disorders, this finding suggests a potential pathological association between chronic neurodegeneration and N proteins [134,135]. Notably, the N protein exhibits critical function in impeding tumor proliferation and metastasis in colon and kidney cancers, an effect mediated through both its NTD and CTD [136]. It also destabilizes the expression of pyruvate kinase M (PKM), an enzyme that promotes tumor proliferation and metastasis, by interacting with another key regulator, Y-box binding protein 1 (YBX1), through the CTD domain, thereby facilitating the G3BP1-mediated recruitment of PKM mRNA into SGs [136]. Additionally, the N protein exacerbates pre-existing cognitive impairment in mice, suggesting that individuals with underlying neurodegenerative diseases may experience a worsening of their condition following exposure to SARS-CoV-2 and the development of COVID-19 [95].

4. Conclusions and Future Work

In summary, the N protein is essential to the SARS-CoV-2 viral life cycle, including viral replication, translation, and assembly, and contributes to viral pathogenesis by interacting with various immune regulatory pathways. Compared to other SARS-CoV-2 viral proteins, the N protein functions as an immunodominant driver due to its abundant and persistent presence in host cells, along with its extensive interactions with multiple immune regulatory pathways. As a result, it plays a central role in both acute immune activation and the development of chronic post-acute sequelae.
Further elucidation of the comprehensive mechanisms of N protein in viral replication and SARS-CoV-2 pathogenesis is requisite for reducing virulence and controlling acute disease progression. Remarkably, N protein also interacts with various pre-existing inflammatory diseases, including diabetes, obesity, and cancer. Due to its prolonged presence and immunogenicity in infected hosts, the N protein is also implicated in long COVID. Unraveling the mechanisms underlying the persistent effects of PASC will provide a crucial foundation for the development of advanced antiviral therapies. There is also an urgent need to understand the long-term immune responses to N protein and to identify strategies for eliminating its persistent presence, not only in the respiratory system but also in other susceptible tissues.
Although mRNA-based COVID-19 vaccines targeting the S gene have been successfully deployed worldwide and proven to be effective, the evolution of SARS-CoV-2 due to immune escape mutations makes it challenging to maintain adequate herd immunity. Given that N protein interacts with various host factors to drive viral pathogenesis and exhibits higher sequence conservation than the S protein, studies using various vaccine platforms have shown the usefulness of including N protein in vaccine composition to broaden adaptive immune responses [137,138]. A proof-of-concept study has also shown that an anti-N monoclonal antibody (mAb) can block complement hyperactivation induced by the N protein [139]. Another study demonstrated that anti-N mAb reduced viral load in the lungs of infected mice when given as a prophylactic treatment [140]. Thus, further work could also focus on developing new therapeutic treatments for COVID-19 by using mAbs, nanobodies, or small molecules to target the N protein.

Author Contributions

Conceptualization, Y.C. and Y.-J.T.; validation, Y.C., Y.W. and Y.-J.T.; writing—original draft preparation, Y.C.; writing—review and editing, Y.C., Y.W., D.H. and Y.-J.T.; visualization, Y.C.; supervision, D.H. and Y.-J.T.; project administration, Y.-J.T. and D.H.; funding acquisition, Y.-J.T. All authors have read and agreed to the published version of the manuscript.

Funding

The Ministry of Education (Singapore) provided financial support for this project by providing a scholarship to Yujia Cao and Tier 2 funding (grant no: T2EP30121-0012) to support Yaju Wang. Dejian Huang is grateful to the Biomedical and Health Technology Platform, National University of Singapore (Suzhou) Research Institute, for financial support.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
AP3B1Adaptor-related protein complex 3 subunit beta 1
ASCApoptosis-associated speck-like protein containing CARD
ASCC3Activating signal co-integrator 1 complex subunit 3
CFTRCystic fibrosis transmembrane conductance regulator
COVID-19Coronavirus disease 2019
CoVsCoronaviruses
CRPC-reactive protein
CTDC-terminal domain
dsRNADouble-stranded RNA
EEnvelope protein
ERGICEndoplasmic reticulum–Golgi intermediate compartment
ERK1/2Extracellular signal-regulated kinases 1 and 2
G3BP1GTPase-activating protein-binding protein 1
GADD34Growth arrest and DNA damage-inducible 34
GSDMDGasdermin D
GSK-3Glycogen synthase kinase-
HCoVHuman coronavirus
HDAC6Histone deacetylases 6
HERC5HECT and RLD domain containing E3 ubiquitin protein ligase 5
HLAHuman leukocyte antigen
ICUIntensive care unit
IDRsIntrinsically disordered regions
IFN-1Type I interferon
IgGImmunoglobulin G
IgMImmunoglobulin M
IKKαIκB kinase-α
ILInterleukin
IMPDH2Inosine monophosphate dehydrogenase 2
IP-10IFN-gamma-inducible protein 10 kD
IRFInterferon regulatory factor
ISG15IFN-stimulated gene 15
JAK-STATJanus kinase-signal transducer and activator of transcription
KPNAKaryopherin alpha
KPNB1Karyopherin beta 1
LKRLinking region
LLPSLiquid–liquid phase separation
MMembrane protein
MAPKMitogen-activated protein kinase
MASP-2Mannan-binding lectin serine protease 2
MAVSMitochondrial antiviral signaling
MDA5Melanoma differentiation-associated gene 5
MERS-CoVMiddle East respiratory syndrome coronavirus
MHVMouse Hepatitis Virus
MIS-CMultisystem inflammatory syndrome
NNucleocapsid
NF-κBNuclear factor kappa B
NFKB1NF-κB subunit 1
NFKBIANF-κB inhibitor alpha
NLRP3NLR family pyrin domain containing-3
NLRsNOD like receptors
NMDNonsense-mediated mRNA decay
NODNucleotide-binding oligomerization domain proteins
NSPsNonstructural proteins
NTDN-terminal domain
ORFsOpen reading frames
PAMPsPathogen-associated molecular patterns
PASCPost-acute sequelae of SARS-CoV-2 infection
PCTProcalcitonin
PDE4Phosphodiesterase 4
PKMPyruvate kinase M
PLproPapain-like protease
poly(I:C)Polyinosinic: polycytidylic acid
PRRsPattern recognition receptors
PTX3Pentraxin 3
RAGEGlycation end products
RANRas-related nuclear protein
RdRpRNA polymerase
RIG-IRetinoic acid-inducible gene I
RISCRNA-induced silencing complex
RLRsRIG-I-like receptors
RNAiRNA interference
RNPRibonucleoprotein
SSpike protein
SAASerum amyloid A
SARS-CoV-2Severe acute respiratory syndrome coronavirus 2
SARSr-CoVSARS-related coronavirus
SCAPSREBPs cleavage-activating protein
SGK1Serum glucocorticoid regulated kinase 1
SGsStress granules
siRNASmall interfering RNAs
SLStem-loop
SNX8Sorting nexin 8
SRSer/Arg
SREBPsSterol regulatory element-binding proteins
SUMOSmall ubiquitin-like modifiers
T1DType 1 diabetes
T2DType 2 diabetes
TBK1Tank-binding kinase 1
TCRT cell receptor
TDP-43Transactive response-binding protein 43 kDa
Th1T helper type 1
TLR2Toll-like receptor 2
TMAOTrimethylamine N-oxide
TNFTumor necrosis factor
TNFR2Tumor necrosis factor receptor 2
TRIM25Tripartite motif protein 25
TRIM6Tripartite motif protein 6
UBC9Ubiquitin-conjugating enzyme 9
UPF1UP-frameshift-1
VOCsVariants of concern
YBX1Y-box binding protein 1

References

  1. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef]
  2. De Wit, E.; Van Doremalen, N.; Falzarano, D.; Munster, V.J. SARS and MERS: Recent insights into emerging coronaviruses. Nat. Rev. Microbiol. 2016, 14, 523–534. [Google Scholar] [CrossRef]
  3. Zmasek, C.M.; Lefkowitz, E.J.; Niewiadomska, A.; Scheuermann, R.H. Genomic evolution of the Coronaviridae family. Virology 2022, 570, 123–133. [Google Scholar] [CrossRef]
  4. Zandi, M.; Shafaati, M.; Kalantar-Neyestanaki, D.; Pourghadamyari, H.; Fani, M.; Soltani, S.; Kaleji, H.; Abbasi, S. The role of SARS-CoV-2 accessory proteins in immune evasion. Biomed. Pharmacother. 2022, 156, 113889. [Google Scholar] [CrossRef]
  5. Wu, W.; Cheng, Y.; Zhou, H.; Sun, C.; Zhang, S. The SARS-CoV-2 nucleocapsid protein: Its role in the viral life cycle, structure and functions, and use as a potential target in the development of vaccines and diagnostics. Virol. J. 2023, 20, 6. [Google Scholar] [CrossRef]
  6. Schiavina, M.; Pontoriero, L.; Uversky, V.N.; Felli, I.C.; Pierattelli, R. The highly flexible disordered regions of the SARS-CoV-2 nucleocapsid N protein within the 1–248 residue construct: Sequence-specific resonance assignments through NMR. Biomol. NMR Assign. 2021, 15, 219–227. [Google Scholar] [CrossRef]
  7. Whittaker, G.R.; Daniel, S.; Millet, J.K. Coronavirus entry: How we arrived at SARS-CoV-2. Curr. Opin. Virol. 2021, 47, 113–120. [Google Scholar] [CrossRef]
  8. Baggen, J.; Vanstreels, E.; Jansen, S.; Daelemans, D. Cellular host factors for SARS-CoV-2 infection. Nat. Microbiol. 2021, 6, 1219–1232. [Google Scholar] [CrossRef]
  9. Gorkhali, R.; Koirala, P.; Rijal, S.; Mainali, A.; Baral, A.; Bhattarai, H.K. Structure and function of major SARS-CoV-2 and SARS-CoV proteins. Bioinform. Biol. Insights 2021, 15, 11779322211025876. [Google Scholar] [CrossRef]
  10. Zeng, W.; Liu, G.; Ma, H.; Zhao, D.; Yang, Y.; Liu, M.; Mohammed, A.; Zhao, C.; Yang, Y.; Xie, J. Biochemical characterization of SARS-CoV-2 nucleocapsid protein. Biochem. Biophys. Res. Commun. 2020, 527, 618–623. [Google Scholar] [CrossRef]
  11. Zhang, X.; Zheng, R.; Li, Z.; Ma, J. Liquid-liquid phase separation in viral function. J. Mol. Biol. 2023, 435, 167955. [Google Scholar] [CrossRef]
  12. Chen, S.-C.; Xu, C.-T.; Chang, C.-F.; Yang, C.-S.; Lin, P.-H.; Liu, W.-M.; Chen, Y.; Yu, C.-H. Characterization of the binding features between SARS-CoV-2 5’-proximal transcripts of genomic RNA and nucleocapsid proteins. RNA Biol. 2025, 22, 1–16. [Google Scholar] [CrossRef]
  13. Zhao, D.; Xu, W.; Zhang, X.; Wang, X.; Ge, Y.; Yuan, E.; Xiong, Y.; Wu, S.; Li, S.; Wu, N. Understanding the phase separation characteristics of nucleocapsid protein provides a new therapeutic opportunity against SARS-CoV-2. Protein Cell 2021, 12, 734–740. [Google Scholar] [CrossRef]
  14. Zhao, M.; Yu, Y.; Sun, L.-M.; Xing, J.-Q.; Li, T.; Zhu, Y.; Wang, M.; Yu, Y.; Xue, W.; Xia, T. GCG inhibits SARS-CoV-2 replication by disrupting the liquid phase condensation of its nucleocapsid protein. Nat. Commun. 2021, 12, 2114. [Google Scholar] [CrossRef]
  15. Johnson, B.A.; Zhou, Y.; Lokugamage, K.G.; Vu, M.N.; Bopp, N.; Crocquet-Valdes, P.A.; Kalveram, B.; Schindewolf, C.; Liu, Y.; Scharton, D. Nucleocapsid mutations in SARS-CoV-2 augment replication and pathogenesis. PLoS Pathog. 2022, 18, e1010627. [Google Scholar] [CrossRef]
  16. Cong, Y.; Ulasli, M.; Schepers, H.; Mauthe, M.; V’kovski, P.; Kriegenburg, F.; Thiel, V.; de Haan, C.A.; Reggiori, F. Nucleocapsid protein recruitment to replication-transcription complexes plays a crucial role in coronaviral life cycle. J. Virol. 2020, 94, e01925-19. [Google Scholar] [CrossRef]
  17. Yaron, T.M.; Heaton, B.E.; Levy, T.M.; Johnson, J.L.; Jordan, T.X.; Cohen, B.M.; Kerelsky, A.; Lin, T.-Y.; Liberatore, K.M.; Bulaon, D.K. Host protein kinases required for SARS-CoV-2 nucleocapsid phosphorylation and viral replication. Sci. Signal. 2022, 15, eabm0808. [Google Scholar] [CrossRef]
  18. Wu, C.-H.; Chen, P.-J.; Yeh, S.-H. Nucleocapsid phosphorylation and RNA helicase DDX1 recruitment enables coronavirus transition from discontinuous to continuous transcription. Cell Host Microbe 2014, 16, 462–472. [Google Scholar] [CrossRef]
  19. Han, Y.; Zhou, H.; Liu, C.; Wang, W.; Qin, Y.; Chen, M. SARS-CoV-2 N protein coordinates viral particle assembly through multiple domains. J. Virol. 2024, 98, e01036-24. [Google Scholar] [CrossRef]
  20. Dutta, M.; Su, Y.; Plescia, C.B.; Voth, G.A.; Stahelin, R.V. The SARS-CoV-2 nucleoprotein associates with anionic lipid membranes. J. Biol. Chem. 2024, 300, 107456. [Google Scholar] [CrossRef]
  21. Zhou, J.; Zhou, Y.; Wei, X.F.; Fan, L.; Gao, X.; Li, Y.; Wu, Y.; Feng, W.; Shen, X.; Liu, L. TRIM6 facilitates SARS-CoV-2 proliferation by catalyzing the K29-typed ubiquitination of NP to enhance the ability to bind viral genomes. J. Med. Virol. 2024, 96, e29531. [Google Scholar] [CrossRef]
  22. Dang, M.; Song, J. CTD of SARS-CoV-2 N protein is a cryptic domain for binding ATP and nucleic acid that interplay in modulating phase separation. Protein Sci. 2022, 31, 345–356. [Google Scholar] [CrossRef]
  23. Brennan, K.; Bowie, A.G. Activation of host pattern recognition receptors by viruses. Curr. Opin. Microbiol. 2010, 13, 503–507. [Google Scholar] [CrossRef]
  24. Kato, H.; Takeuchi, O.; Sato, S.; Yoneyama, M.; Yamamoto, M.; Matsui, K.; Uematsu, S.; Jung, A.; Kawai, T.; Ishii, K.J. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature 2006, 441, 101–105. [Google Scholar] [CrossRef]
  25. Wang, M.; Zhao, Y.; Liu, J.; Li, T. SARS-CoV-2 modulation of RIG-I-MAVS signaling: Potential mechanisms of impairment on host antiviral immunity and therapeutic approaches. MedComm–Future Med. 2022, 1, e29. [Google Scholar] [CrossRef]
  26. Aoshi, T.; Koyama, S.; Kobiyama, K.; Akira, S.; Ishii, K.J. Innate and adaptive immune responses to viral infection and vaccination. Curr. Opin. Virol. 2011, 1, 226–232. [Google Scholar] [CrossRef]
  27. Hu, Y.; Li, W.; Gao, T.; Cui, Y.; Jin, Y.; Li, P.; Ma, Q.; Liu, X.; Cao, C. The severe acute respiratory syndrome coronavirus nucleocapsid inhibits type I interferon production by interfering with TRIM25-mediated RIG-I ubiquitination. J. Virol. 2017, 91, e02143-16. [Google Scholar] [CrossRef]
  28. Oh, S.J.; Shin, O.S. SARS-CoV-2 nucleocapsid protein targets RIG-I-like receptor pathways to inhibit the induction of interferon response. Cells 2021, 10, 530. [Google Scholar] [CrossRef]
  29. Chen, K.; Xiao, F.; Hu, D.; Ge, W.; Tian, M.; Wang, W.; Pan, P.; Wu, K.; Wu, J. SARS-CoV-2 nucleocapsid protein interacts with RIG-I and represses RIG-mediated IFN-β production. Viruses 2020, 13, 47. [Google Scholar] [CrossRef]
  30. Li, Y.; Li, M.; Xiao, H.; Liao, F.; Shen, M.; Ge, W.; Ou, J.; Liu, Y.; Chen, L.; Zhao, Y. The R203M and D377Y mutations of the nucleocapsid protein promote SARS-CoV-2 infectivity by impairing RIG-I-mediated antiviral signaling. PLoS Pathog. 2025, 21, e1012886. [Google Scholar] [CrossRef]
  31. Zheng, Z.-Q.; Wang, S.-Y.; Xu, Z.-S.; Fu, Y.-Z.; Wang, Y.-Y. SARS-CoV-2 nucleocapsid protein impairs stress granule formation to promote viral replication. Cell Discov. 2021, 7, 38. [Google Scholar] [CrossRef]
  32. Zheng, Y.; Deng, J.; Han, L.; Zhuang, M.-W.; Xu, Y.; Zhang, J.; Nan, M.-L.; Xiao, Y.; Zhan, P.; Liu, X. SARS-CoV-2 NSP5 and N protein counteract the RIG-I signaling pathway by suppressing the formation of stress granules. Signal Transduct. Target. Ther. 2022, 7, 22. [Google Scholar] [CrossRef]
  33. Kim, S.S.-Y.; Sze, L.; Lam, K.-P. The stress granule protein G3BP1 binds viral dsRNA and RIG-I to enhance interferon-β response. J. Biol. Chem. 2019, 294, 6430–6438. [Google Scholar] [CrossRef]
  34. Somasekharan, S.P.; Gleave, M. SARS-CoV-2 nucleocapsid protein interacts with immunoregulators and stress granules and phase separates to form liquid droplets. FEBS Lett. 2021, 595, 2872–2896. [Google Scholar] [CrossRef]
  35. Mukherjee, A.; Lo, M.; Chandra, P.; Datta Chaudhuri, R.; De, P.; Dutta, S.; Chawla-Sarkar, M. SARS-CoV-2 nucleocapsid protein promotes self-deacetylation by inducing HDAC6 to facilitate viral replication. Virol. J. 2024, 21, 186. [Google Scholar] [CrossRef]
  36. Mulloy, R.P.; Evseev, D.; Bui-Marinos, M.P.; Sharlin, N.; Corcoran, J.A. A truncated SARS-CoV-2 nucleocapsid protein enhances virus fitness by evading antiviral responses. bioRxiv 2025. [Google Scholar] [CrossRef]
  37. Liu, J.; Guan, G.; Wu, C.; Wang, B.; Chu, K.; Zhang, X.; He, S.; Zhang, N.; Yang, G.; Jin, Z. SARS-CoV-2 Nucleocapsid Protein Antagonizes GADD34-Mediated Innate Immune Pathway through Atypical Foci. Molecules 2024, 29, 4792. [Google Scholar] [CrossRef]
  38. Wang, S.; Dai, T.; Qin, Z.; Pan, T.; Chu, F.; Lou, L.; Zhang, L.; Yang, B.; Huang, H.; Lu, H. Targeting liquid–liquid phase separation of SARS-CoV-2 nucleocapsid protein promotes innate antiviral immunity by elevating MAVS activity. Nat. Cell Biol. 2021, 23, 718–732. [Google Scholar] [CrossRef]
  39. Lu, X.; Pan, J.; Tao, J.; Guo, D. SARS-CoV nucleocapsid protein antagonizes IFN-β response by targeting initial step of IFN-β induction pathway, and its C-terminal region is critical for the antagonism. Virus Genes 2011, 42, 37–45. [Google Scholar] [CrossRef]
  40. Zotta, A.; Hooftman, A.; O’Neill, L.A. SARS-CoV-2 targets MAVS for immune evasion. Nat. Cell Biol. 2021, 23, 682–683. [Google Scholar] [CrossRef]
  41. Choi, G.W.; Lee, Y.; Yun, M.; Kang, J.; Lee, S.-B. Formation of SUMO3-conjugated chains of MAVS induced by poly (dA:dT), a ligand of RIG-I, enhances the aggregation of MAVS that drives the secretion of interferon-β in human keratinocytes. Biochem. Biophys. Res. Commun. 2020, 522, 939–944. [Google Scholar] [CrossRef]
  42. Huang, C.; Yin, Y.; Pan, P.; Huang, Y.; Chen, S.; Chen, J.; Wang, J.; Xu, G.; Tao, X.; Xiao, X. The Interaction between SARS-CoV-2 Nucleocapsid Protein and UBC9 Inhibits MAVS Ubiquitination by Enhancing Its SUMOylation. Viruses 2023, 15, 2304. [Google Scholar] [CrossRef]
  43. Ren, J.; Wang, S.; Zong, Z.; Pan, T.; Liu, S.; Mao, W.; Huang, H.; Yan, X.; Yang, B.; He, X. TRIM28-mediated nucleocapsid protein SUMOylation enhances SARS-CoV-2 virulence. Nat. Commun. 2024, 15, 244. [Google Scholar] [CrossRef]
  44. Mao, S.; Cai, X.; Niu, S.; Wei, J.; Jiang, N.; Deng, H.; Wang, W.; Zhang, J.; Shen, S.; Ma, Y. TRIM21 promotes ubiquitination of SARS-CoV-2 nucleocapsid protein to regulate innate immunity. J. Med. Virol. 2023, 95, e28719. [Google Scholar] [CrossRef]
  45. Shajahan, A.; Pepi, L.E.; Rouhani, D.S.; Heiss, C.; Azadi, P. Glycosylation of SARS-CoV-2: Structural and functional insights. Anal. Bioanal. Chem. 2021, 413, 7179–7193. [Google Scholar] [CrossRef]
  46. Rump, A.; Risti, R.; Kristal, M.-L.; Reut, J.; Syritski, V.; Lookene, A.; Boudinot, S.R. Dual ELISA using SARS-CoV-2 nucleocapsid protein produced in E. coli and CHO cells reveals epitope masking by N-glycosylation. Biochem. Biophys. Res. Commun. 2021, 534, 457–460. [Google Scholar] [CrossRef]
  47. Nilsson-Payant, B.E.; Uhl, S.; Grimont, A.; Doane, A.S.; Cohen, P.; Patel, R.S.; Higgins, C.A.; Acklin, J.A.; Bram, Y.; Chandar, V. The NF-κB transcriptional footprint is essential for SARS-CoV-2 replication. J. Virol. 2021, 95, e0125721. [Google Scholar] [CrossRef]
  48. Qian, Y.; Lei, T.; Patel, P.S.; Lee, C.H.; Monaghan-Nichols, P.; Xin, H.-B.; Qiu, J.; Fu, M. Direct activation of endothelial cells by SARS-CoV-2 nucleocapsid protein is blocked by Simvastatin. J. Virol. 2021, 95, e0139621. [Google Scholar] [CrossRef]
  49. Wang, M.; Valadez-Ingersoll, M.; Gilmore, T.D. Control of nuclear localization of the nucleocapsid protein of SARS-CoV-2. Virology 2024, 600, 110232. [Google Scholar] [CrossRef]
  50. Lu, Y.; Ye, Z.; Liu, X.; Zhou, L.; Ding, X.; Hou, Y. Role of SARS-CoV-2 nucleocapsid protein in affecting immune cells and insights on its molecular mechanisms. Exp. Ther. Med. 2023, 26, 504. [Google Scholar] [CrossRef]
  51. Wang, Y.C.; Tsai, C.-H.; Wang, Y.-C.; Yen, L.-C.; Chang, Y.-W.; Sun, J.-R.; Lin, T.-Y.; Chiu, C.-H.; Chao, Y.-C.; Chang, F.-Y. SARS-CoV-2 nucleocapsid protein, rather than spike protein, triggers a cytokine storm originating from lung epithelial cells in patients with COVID-19. Infection 2024, 52, 955–983. [Google Scholar] [CrossRef]
  52. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.-C.; Uhl, S.; Hoagland, D.; Møller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 2020, 181, 1036–1045.e9. [Google Scholar] [CrossRef]
  53. Xia, J.; Tang, W.; Wang, J.; Lai, D.; Xu, Q.; Huang, R.; Hu, Y.; Gong, X.; Fan, J.; Shu, Q. SARS-CoV-2 N protein induces acute lung injury in mice via NF-ĸB activation. Front. Immunol. 2021, 12, 791753. [Google Scholar] [CrossRef]
  54. Wu, Y.; Ma, L.; Cai, S.; Zhuang, Z.; Zhao, Z.; Jin, S.; Xie, W.; Zhou, L.; Zhang, L.; Zhao, J. RNA-induced liquid phase separation of SARS-CoV-2 nucleocapsid protein facilitates NF-κB hyper-activation and inflammation. Signal Transduct. Target. Ther. 2021, 6, 167. [Google Scholar] [CrossRef]
  55. Xia, J.; Wang, J.; Ying, L.; Huang, R.; Zhang, K.; Zhang, R.; Tang, W.; Xu, Q.; Lai, D.; Zhang, Y. RAGE is a receptor for SARS-CoV-2 N protein and mediates N protein–induced acute lung injury. Am. J. Respir. Cell Mol. Biol. 2023, 69, 508–520. [Google Scholar] [CrossRef]
  56. Yue, Z.; Zhang, X.; Gu, Y.; Liu, Y.; Lan, L.-M.; Liu, Y.; Li, Y.; Yang, G.; Wan, P.; Chen, X. Regulation and functions of the NLRP3 inflammasome in RNA virus infection. Front. Cell. Infect. Microbiol. 2024, 13, 1309128. [Google Scholar] [CrossRef]
  57. Pan, P.; Shen, M.; Yu, Z.; Ge, W.; Chen, K.; Tian, M.; Xiao, F.; Wang, Z.; Wang, J.; Jia, Y. SARS-CoV-2 N protein promotes NLRP3 inflammasome activation to induce hyperinflammation. Nat. Commun. 2021, 12, 4664. [Google Scholar] [CrossRef]
  58. Ma, J.; Zhu, F.; Zhao, M.; Shao, F.; Yu, D.; Ma, J.; Zhang, X.; Li, W.; Qian, Y.; Zhang, Y. SARS-CoV-2 nucleocapsid suppresses host pyroptosis by blocking Gasdermin D cleavage. EMBO J. 2021, 40, e108249. [Google Scholar] [CrossRef]
  59. Tan, F.L.; Yin, J.Q. RNAi, a new therapeutic strategy against viral infection. Cell Res. 2004, 14, 460–466. [Google Scholar] [CrossRef]
  60. van Rij, R.P.; Andino, R. The silent treatment: RNAi as a defense against virus infection in mammals. Trends Biotechnol. 2006, 24, 186–193. [Google Scholar] [CrossRef]
  61. Mu, J.; Xu, J.; Zhang, L.; Shu, T.; Wu, D.; Huang, M.; Ren, Y.; Li, X.; Geng, Q.; Xu, Y. SARS-CoV-2-encoded nucleocapsid protein acts as a viral suppressor of RNA interference in cells. Sci. China Life Sci. 2020, 63, 1413–1416. [Google Scholar] [CrossRef]
  62. Cui, L.; Wang, H.; Ji, Y.; Yang, J.; Xu, S.; Huang, X.; Wang, Z.; Qin, L.; Tien, P.; Zhou, X. The nucleocapsid protein of coronaviruses acts as a viral suppressor of RNA silencing in mammalian cells. J. Virol. 2015, 89, 9029–9043. [Google Scholar] [CrossRef]
  63. Nuccetelli, V.; Mghezzi-Habellah, M.; Deymier, S.; Roisin, A.; Gérard-Baraggia, F.; Rocchi, C.; Coureux, P.-D.; Gouet, P.; Cimarelli, A.; Mocquet, V. The SARS-CoV-2 nucleocapsid protein interferes with the full enzymatic activation of UPF1 and its interaction with UPF2. Nucleic Acids Res. 2025, 53, gkaf010. [Google Scholar] [CrossRef]
  64. Jiang, P.; Dai, Z.; Yang, C.; Ding, L.; Li, S.; Xu, X.; Cheng, C.; Wang, J.; Liu, S. CFTR Inhibitors Display Antiviral Activity against Herpes Simplex Virus. Viruses 2024, 16, 1308. [Google Scholar] [CrossRef]
  65. Chen, L.; Guan, W.-J.; Qiu, Z.-E.; Xu, J.-B.; Bai, X.; Hou, X.-C.; Sun, J.; Qu, S.; Huang, Z.-X.; Lei, T.-L. SARS-CoV-2 nucleocapsid protein triggers hyperinflammation via protein-protein interaction-mediated intracellular Cl accumulation in respiratory epithelium. Signal Transduct. Target. Ther. 2022, 7, 255. [Google Scholar] [CrossRef]
  66. Dzimianski, J.V.; Scholte, F.E.; Bergeron, É.; Pegan, S.D. ISG15: It’s complicated. J. Mol. Biol. 2019, 431, 4203–4216. [Google Scholar] [CrossRef]
  67. Zhu, J.; Liu, G.; Sayyad, Z.; Goins, C.M.; Stauffer, S.R.; Gack, M.U. ISGylation of the SARS-CoV-2 N protein by HERC5 impedes N oligomerization and thereby viral RNA synthesis. J. Virol. 2024, 98, e00869-24. [Google Scholar] [CrossRef]
  68. Rhamadianti, A.F.; Abe, T.; Tanaka, T.; Ono, C.; Katayama, H.; Makino, Y.; Deng, L.; Matsui, C.; Moriishi, K.; Shima, F. SARS-CoV-2 papain-like protease inhibits ISGylation of the viral nucleocapsid protein to evade host anti-viral immunity. J. Virol. 2024, 98, e00855-24. [Google Scholar] [CrossRef]
  69. Bang, W.; Kim, J.; Seo, K.; Lee, J.; Han, J.H.; Park, D.; Cho, J.H.; Shin, D.; Kim, K.-H.; Song, M.J. Suppression of SARS-CoV-2 nucleocapsid protein dimerization by ISGylation and its counteraction by viral PLpro. Front. Microbiol. 2024, 15, 1490944. [Google Scholar] [CrossRef]
  70. Fujita, T. Evolution of the lectin–complement pathway and its role in innate immunity. Nat. Rev. Immunol. 2002, 2, 346–353. [Google Scholar] [CrossRef]
  71. Gao, T.; Zhu, L.; Liu, H.; Zhang, X.; Wang, T.; Fu, Y.; Li, H.; Dong, Q.; Hu, Y.; Zhang, Z. Highly pathogenic coronavirus N protein aggravates inflammation by MASP-2-mediated lectin complement pathway overactivation. Signal Transduct. Target. Ther. 2022, 7, 318. [Google Scholar] [CrossRef]
  72. Kocsis, A.; Bartus, D.; Hirsch, E.; Józsi, M.; Hajdú, I.; Dobó, J.; Balczer, J.; Pál, G.; Gál, P. SARS-CoV-2 Nucleocapsid Protein Is Not Responsible for Over-Activation of Complement Lectin Pathway. Int. J. Mol. Sci. 2024, 25, 7343. [Google Scholar] [CrossRef]
  73. Bally, I.; Drumont, G.; Rossi, V.; Guseva, S.; Botova, M.; Reiser, J.-B.; Thépaut, M.; Dergan Dylon, S.; Dumestre-Pérard, C.; Gaboriaud, C. Revisiting the interaction between complement lectin pathway protease MASP-2 and SARS-CoV-2 nucleoprotein. Front. Immunol. 2024, 15, 1419165. [Google Scholar] [CrossRef]
  74. Zhao, Y.; Sui, L.; Wu, P.; Wang, W.; Wang, Z.; Yu, Y.; Hou, Z.; Tan, G.; Liu, Q.; Wang, G. A dual-role of SARS-CoV-2 nucleocapsid protein in regulating innate immune response. Signal Transduct. Target. Ther. 2021, 6, 331. [Google Scholar] [CrossRef]
  75. Reuschl, A.-K.; Thorne, L.G.; Whelan, M.V.; Ragazzini, R.; Furnon, W.; Cowton, V.M.; De Lorenzo, G.; Mesner, D.; Turner, J.L.; Dowgier, G. Evolution of enhanced innate immune suppression by SARS-CoV-2 Omicron subvariants. Nat. Microbiol. 2024, 9, 451–463. [Google Scholar] [CrossRef]
  76. Shekhawat, J.; Gauba, K.; Gupta, S.; Purohit, P.; Mitra, P.; Garg, M.; Misra, S.; Sharma, P.; Banerjee, M. Interleukin-6 perpetrator of the COVID-19 cytokine storm. Indian J. Clin. Biochem. 2021, 36, 440–450. [Google Scholar] [CrossRef]
  77. Karwaciak, I.; Sałkowska, A.; Karaś, K.; Dastych, J.; Ratajewski, M. Nucleocapsid and spike proteins of the coronavirus SARS-CoV-2 induce il6 in monocytes and macrophages—Potential implications for cytokine storm syndrome. Vaccines 2021, 9, 54. [Google Scholar] [CrossRef]
  78. Vora, S.M.; Lieberman, J.; Wu, H. Inflammasome activation at the crux of severe COVID-19. Nat. Rev. Immunol. 2021, 21, 694–703. [Google Scholar] [CrossRef]
  79. Hu, B.; Huang, S.; Yin, L. The cytokine storm and COVID-19. J. Med. Virol. 2021, 93, 250–256. [Google Scholar] [CrossRef]
  80. Chen, L.-D.; Zhang, Z.-Y.; Wei, X.-J.; Cai, Y.-Q.; Yao, W.-Z.; Wang, M.-H.; Huang, Q.-F.; Zhang, X.-B. Association between cytokine profiles and lung injury in COVID-19 pneumonia. Respir. Res. 2020, 21, 201. [Google Scholar] [CrossRef]
  81. McConnell, M.J.; Kawaguchi, N.; Kondo, R.; Sonzogni, A.; Licini, L.; Valle, C.; Bonaffini, P.A.; Sironi, S.; Alessio, M.G.; Previtali, G. Liver injury in COVID-19 and IL-6 trans-signaling-induced endotheliopathy. J. Hepatol. 2021, 75, 647–658. [Google Scholar] [CrossRef]
  82. Wick, K.D.; Leligdowicz, A.; Willmore, A.; Carrillo, S.A.; Ghale, R.; Jauregui, A.; Chak, S.S.; Nguyen, V.; Lee, D.; Jones, C. Plasma SARS-CoV-2 nucleocapsid antigen levels are associated with progression to severe disease in hospitalized COVID-19. Crit. Care 2022, 26, 278. [Google Scholar] [CrossRef]
  83. ACTIV-3/TICO Bamlanivimab Study Group; Lundgren, J.D.; Grund, B.; Barkauskas, C.E.; Holland, T.L.; Gottlieb, R.L.; Sandkovsky, U.; Brown, S.M.; Knowlton, K.U.; Self, W.H. Responses to a neutralizing monoclonal antibody for hospitalized patients with COVID-19 according to baseline antibody and antigen levels: A randomized controlled trial. Ann. Intern. Med. 2022, 175, 234–243. [Google Scholar] [CrossRef]
  84. Thudium, R.F.; Stoico, M.P.; Høgdall, E.; Høgh, J.; Krarup, H.B.; Larsen, M.A.; Madsen, P.H.; Nielsen, S.D.; Ostrowski, S.R.; Palombini, A. Early laboratory diagnosis of COVID-19 by antigen detection in blood samples of the SARS-CoV-2 nucleocapsid protein. J. Clin. Microbiol. 2021, 59, e01001-21. [Google Scholar] [CrossRef]
  85. Favresse, J.; Bayart, J.-L.; David, C.; Dogné, J.-M.; Douxfils, J. Nucleocapsid serum antigen determination in SARS-CoV-2 infected patients using the single molecule array technology and prediction of disease severity. J. Infect. 2022, 84, e4. [Google Scholar] [CrossRef]
  86. Yuan, C.; Ma, Z.; Xie, J.; Li, W.; Su, L.; Zhang, G.; Xu, J.; Wu, Y.; Zhang, M.; Liu, W. The role of cell death in SARS-CoV-2 infection. Signal Transduct. Target. Ther. 2023, 8, 357. [Google Scholar] [CrossRef]
  87. Machhi, J.; Shahjin, F.; Das, S.; Patel, M.; Abdelmoaty, M.M.; Cohen, J.D.; Singh, P.A.; Baldi, A.; Bajwa, N.; Kumar, R. A role for extracellular vesicles in SARS-CoV-2 therapeutics and prevention. J. Neuroimmune Pharmacol. 2021, 16, 270–288. [Google Scholar] [CrossRef]
  88. Perna, F.; Bruzzaniti, S.; Piemonte, E.; Maddaloni, V.; Atripaldi, L.; Sale, S.; Sanduzzi, A.; Nicastro, C.; Pepe, N.; Bifulco, M. Serum levels of SARS-CoV-2 nucleocapsid antigen associate with inflammatory status and disease severity in COVID-19 patients. Clin. Immunol. 2021, 226, 108720. [Google Scholar] [CrossRef]
  89. Chen, J.; Zhao, S.; Yan, H.; Huang, Y.; Wei, C.; Liu, J.; Sun, J. Plasma SARS-CoV-2 nucleocapsid antigen levels are associated with lung infection and tissue-damage biomarkers. Virus Res. 2025, 356, 199580. [Google Scholar] [CrossRef]
  90. Wang, L. C-reactive protein levels in the early stage of COVID-19. Med. Mal. Infect. 2020, 50, 332–334. [Google Scholar] [CrossRef]
  91. Chen, W.; Zheng, K.I.; Liu, S.; Yan, Z.; Xu, C.; Qiao, Z. Plasma CRP level is positively associated with the severity of COVID-19. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 18. [Google Scholar] [CrossRef]
  92. Stravalaci, M.; Pagani, I.; Paraboschi, E.M.; Pedotti, M.; Doni, A.; Scavello, F.; Mapelli, S.N.; Sironi, M.; Perucchini, C.; Varani, L. Recognition and inhibition of SARS-CoV-2 by humoral innate immunity pattern recognition molecules. Nat. Immunol. 2022, 23, 275–286. [Google Scholar] [CrossRef]
  93. Feitosa, T.A.; de Souza Sá, M.V.; Pereira, V.C.; de Andrade Cavalcante, M.K.; Pereira, V.R.A.; da Costa Armstrong, A.; do Carmo, R.F. Association of polymorphisms in long pentraxin 3 and its plasma levels with COVID-19 severity. Clin. Exp. Med. 2023, 23, 1225–1233. [Google Scholar] [CrossRef]
  94. Oprinca, G.C.; Mohor, C.-I.; Bereanu, A.-S.; Oprinca-Muja, L.-A.; Bogdan-Duică, I.; Fleacă, S.R.; Hașegan, A.; Diter, A.; Boeraș, I.; Cristian, A.N. Detection of SARS-CoV-2 Viral Genome and Viral Nucleocapsid in Various Organs and Systems. Int. J. Mol. Sci. 2024, 25, 5755. [Google Scholar] [CrossRef]
  95. Orsini, F.; Bosica, M.; Martucci, A.; De Paola, M.; Comolli, D.; Pascente, R.; Forloni, G.; Fraser, P.E.; Arancio, O.; Fioriti, L. SARS-CoV-2 Nucleocapsid Protein Induces Tau Pathological Changes That Can Be Counteracted by SUMO2. Int. J. Mol. Sci. 2024, 25, 7169. [Google Scholar] [CrossRef]
  96. Low, Z.Y.; Zabidi, N.Z.; Yip, A.J.W.; Puniyamurti, A.; Chow, V.T.; Lal, S.K. SARS-CoV-2 non-structural proteins and their roles in host immune evasion. Viruses 2022, 14, 1991. [Google Scholar] [CrossRef]
  97. Demmer, R.T.; Wu, C.; Kim, J.S.; Sun, Y.; Balte, P.; Cushman, M.; Boyle, R.; Tracy, R.P.; Styer, L.M.; Bell, T.D. Demographic and Clinical Factors Associated With SARS-CoV-2 Anti-Nucleocapsid Antibody Response Among Previously Infected US Adults: The C4R Study. Open Forum Infect. Dis. 2025, 12, ofaf123. [Google Scholar] [CrossRef]
  98. Sokal, A.; Chappert, P.; Barba-Spaeth, G.; Roeser, A.; Fourati, S.; Azzaoui, I.; Vandenberghe, A.; Fernandez, I.; Meola, A.; Bouvier-Alias, M. Maturation and persistence of the anti-SARS-CoV-2 memory B cell response. Cell 2021, 184, 1201–1213.E14. [Google Scholar] [CrossRef]
  99. Zhou, P.; Yang, X.-L.; Wang, X.-G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.-R.; Zhu, Y.; Li, B.; Huang, C.-L. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef]
  100. Koerber, N.; Priller, A.; Yazici, S.; Bauer, T.; Cheng, C.-C.; Mijočević, H.; Wintersteller, H.; Jeske, S.; Vogel, E.; Feuerherd, M. Dynamics of spike-and nucleocapsid specific immunity during long-term follow-up and vaccination of SARS-CoV-2 convalescents. Nat. Commun. 2022, 13, 153. [Google Scholar] [CrossRef]
  101. Chansaenroj, J.; Yorsaeng, R.; Posuwan, N.; Puenpa, J.; Wanlapakorn, N.; Sudhinaraset, N.; Sripramote, M.; Chalongviriyalert, P.; Jirajariyavej, S.; Kiatpanabhikul, P. Long-term specific IgG response to SARS-CoV-2 nucleocapsid protein in recovered COVID-19 patients. Sci. Rep. 2021, 11, 23216. [Google Scholar] [CrossRef]
  102. Chia, W.N.; Zhu, F.; Ong, S.W.X.; Young, B.E.; Fong, S.-W.; Le Bert, N.; Tan, C.W.; Tiu, C.; Zhang, J.; Tan, S.Y. Dynamics of SARS-CoV-2 neutralising antibody responses and duration of immunity: A longitudinal study. Lancet Microbe 2021, 2, e240–e249. [Google Scholar] [CrossRef]
  103. Eser, T.M.; Baranov, O.; Huth, M.; Ahmed, M.I.; Deák, F.; Held, K.; Lin, L.; Pekayvaz, K.; Leunig, A.; Nicolai, L. Nucleocapsid-specific T cell responses associate with control of SARS-CoV-2 in the upper airways before seroconversion. Nat. Commun. 2023, 14, 2952. [Google Scholar] [CrossRef]
  104. Peluso, M.J.; Deitchman, A.N.; Torres, L.; Iyer, N.S.; Munter, S.E.; Nixon, C.C.; Donatelli, J.; Thanh, C.; Takahashi, S.; Hakim, J. Long-term SARS-CoV-2-specific immune and inflammatory responses in individuals recovering from COVID-19 with and without post-acute symptoms. Cell Rep. 2021, 36, 109518. [Google Scholar] [CrossRef]
  105. Taus, E.; Hofmann, C.; Ibarrondo, F.J.; Hausner, M.A.; Fulcher, J.A.; Krogstad, P.; Ferbas, K.G.; Tobin, N.H.; Rimoin, A.W.; Aldrovandi, G.M. Dominant CD8+ T cell nucleocapsid targeting in SARS-CoV-2 infection and broad spike targeting from vaccination. Front. Immunol. 2022, 13, 835830. [Google Scholar] [CrossRef]
  106. López-Muñoz, A.D.; Kosik, I.; Holly, J.; Yewdell, J.W. Cell surface SARS-CoV-2 nucleocapsid protein modulates innate and adaptive immunity. Sci. Adv. 2022, 8, eabp9770. [Google Scholar] [CrossRef]
  107. Lineburg, K.E.; Grant, E.J.; Swaminathan, S.; Chatzileontiadou, D.S.; Szeto, C.; Sloane, H.; Panikkar, A.; Raju, J.; Crooks, P.; Rehan, S. CD8+ T cells specific for an immunodominant SARS-CoV-2 nucleocapsid epitope cross-react with selective seasonal coronaviruses. Immunity 2021, 54, 1055–1065.E5. [Google Scholar] [CrossRef]
  108. Nguyen, T.H.; Rowntree, L.C.; Petersen, J.; Chua, B.Y.; Hensen, L.; Kedzierski, L.; van de Sandt, C.E.; Chaurasia, P.; Tan, H.-X.; Habel, J.R. CD8+ T cells specific for an immunodominant SARS-CoV-2 nucleocapsid epitope display high naive precursor frequency and TCR promiscuity. Immunity 2021, 54, 1066–1082.E5. [Google Scholar] [CrossRef]
  109. Nalbandian, A.; Sehgal, K.; Gupta, A.; Madhavan, M.V.; McGroder, C.; Stevens, J.S.; Cook, J.R.; Nordvig, A.S.; Shalev, D.; Sehrawat, T.S. Post-acute COVID-19 syndrome. Nat. Med. 2021, 27, 601–615. [Google Scholar] [CrossRef]
  110. Jia, X.; Cao, S.; Lee, A.S.; Manohar, M.; Sindher, S.B.; Ahuja, N.; Artandi, M.; Blish, C.A.; Blomkalns, A.L.; Chang, I. Anti-nucleocapsid antibody levels and pulmonary comorbid conditions are linked to post–COVID-19 syndrome. JCI Insight 2022, 7, e156713. [Google Scholar] [CrossRef]
  111. Altmann, D.M.; Whettlock, E.M.; Liu, S.; Arachchillage, D.J.; Boyton, R.J. The immunology of long COVID. Nat. Rev. Immunol. 2023, 23, 618–634. [Google Scholar] [CrossRef]
  112. Littlefield, K.M.; Watson, R.O.; Schneider, J.M.; Neff, C.P.; Yamada, E.; Zhang, M.; Campbell, T.B.; Falta, M.T.; Jolley, S.E.; Fontenot, A.P. SARS-CoV-2-specific T cells associate with inflammation and reduced lung function in pulmonary post-acute sequalae of SARS-CoV-2. PloS Pathog. 2022, 18, e1010359. [Google Scholar] [CrossRef]
  113. Schultheiß, C.; Willscher, E.; Paschold, L.; Gottschick, C.; Klee, B.; Henkes, S.-S.; Bosurgi, L.; Dutzmann, J.; Sedding, D.; Frese, T. The IL-1β, IL-6, and TNF cytokine triad is associated with post-acute sequelae of COVID-19. Cell Rep. Med. 2022, 3, 100663. [Google Scholar] [CrossRef]
  114. Patrascu, R.; Dumitru, C.S. Advances in Understanding Inflammation and Tissue Damage: Markers of Persistent Sequelae in COVID-19 Patients. J. Clin. Med. 2025, 14, 1475. [Google Scholar] [CrossRef]
  115. Yin, J.-X.; Agbana, Y.L.; Sun, Z.-S.; Fei, S.-W.; Zhao, H.-Q.; Zhou, X.-N.; Chen, J.-H.; Kassegne, K. Increased interleukin-6 is associated with long COVID-19: A systematic review and meta-analysis. Infect. Dis. Poverty 2023, 12, 43. [Google Scholar] [CrossRef]
  116. Santa Cruz, A.; Mendes-Frias, A.; Azarias-da-Silva, M.; André, S.; Oliveira, A.I.; Pires, O.; Mendes, M.; Oliveira, B.; Braga, M.; Lopes, J.R. Post-acute sequelae of COVID-19 is characterized by diminished peripheral CD8+ β7 integrin+ T cells and anti-SARS-CoV-2 IgA response. Nat. Commun. 2023, 14, 1772. [Google Scholar] [CrossRef]
  117. Grote, K.; Schaefer, A.-C.; Soufi, M.; Ruppert, V.; Linne, U.; Mukund Bhagwat, A.; Szymanski, W.; Graumann, J.; Gercke, Y.; Aldudak, S. Targeting the high-density lipoprotein proteome for the treatment of post-acute sequelae of SARS-CoV-2. Int. J. Mol. Sci. 2024, 25, 4522. [Google Scholar] [CrossRef]
  118. Visvabharathy, L.; Hanson, B.A.; Orban, Z.S.; Lim, P.H.; Palacio, N.M.; Jimenez, M.; Clark, J.R.; Graham, E.L.; Liotta, E.M.; Tachas, G. Neuro-PASC is characterized by enhanced CD4+ and diminished CD8+ T cell responses to SARS-CoV-2 Nucleocapsid protein. Front. Immunol. 2023, 14, 1155770. [Google Scholar] [CrossRef]
  119. Bodansky, A.; Mettelman, R.C.; Sabatino, J.J., Jr.; Vazquez, S.E.; Chou, J.; Novak, T.; Moffitt, K.L.; Miller, H.S.; Kung, A.F.; Rackaityte, E. Molecular mimicry in multisystem inflammatory syndrome in children. Nature 2024, 632, 622–629. [Google Scholar] [CrossRef]
  120. Oprinca, G.-C.; Mohor, C.-I.; Oprinca-Muja, A.; Hașegan, A.; Cristian, A.-N.; Fleacă, S.-R.; Boeraș, I.; Cardoș, R.; Atasie, D.; Mihalache, M. Unveiling the Pathological Mechanisms of Death Induced by SARS-CoV-2 Viral Pneumonia. Microorganisms 2024, 12, 459. [Google Scholar] [CrossRef]
  121. Yan, Y.; Diao, B.; Liu, Y.; Zhang, W.; Wang, G.; Chen, X. Severe acute respiratory syndrome coronavirus 2 nucleocapsid protein in the ocular tissues of a patient previously infected with coronavirus disease 2019. JAMA Ophthalmol. 2020, 138, 1201–1204. [Google Scholar] [CrossRef]
  122. Yokoyama, K.; Sato, Y.; Suimon, Y.; Kase, S. Late onset paediatric dacryoadenitis associated with SARS-CoV-2 confirmed by histological analysis. BMJ Case Rep. CP 2024, 17, e257615. [Google Scholar] [CrossRef]
  123. de Melo, G.D.; Lazarini, F.; Levallois, S.; Hautefort, C.; Michel, V.; Larrous, F.; Verillaud, B.; Aparicio, C.; Wagner, S.; Gheusi, G. COVID-19–related anosmia is associated with viral persistence and inflammation in human olfactory epithelium and brain infection in hamsters. Sci. Transl. Med. 2021, 13, eabf8396. [Google Scholar] [CrossRef]
  124. El-Baky, N.A.; Amara, A.A.; Uversky, V.N.; Redwan, E.M. Intrinsic factors behind long COVID: III. Persistence of SARS-CoV-2 and its components. J. Cell. Biochem. 2024, 125, e30514. [Google Scholar] [CrossRef]
  125. Eltayeb, A.; Al-Sarraj, F.; Alharbi, M.; Albiheyri, R.; Mattar, E.H.; Abu Zeid, I.M.; Bouback, T.A.; Bamagoos, A.; Uversky, V.N.; Rubio-Casillas, A. Intrinsic factors behind long COVID: IV. Hypothetical roles of the SARS-CoV-2 nucleocapsid protein and its liquid–liquid phase separation. J. Cell. Biochem. 2024, 125, e30530. [Google Scholar] [CrossRef]
  126. Rizzo, R.; Neri, L.M.; Simioni, C.; Bortolotti, D.; Occhionorelli, S.; Zauli, G.; Secchiero, P.; Semprini, C.M.; Laface, I.; Sanz, J.M. SARS-CoV-2 nucleocapsid protein and ultrastructural modifications in small bowel of a 4-week-negative COVID-19 patient. Clin. Microbiol. Infect. 2021, 27, 936. [Google Scholar] [CrossRef]
  127. Falagas, M.E.; Kompoti, M. Obesity and infection. Lancet Infect. Dis. 2006, 6, 438–446. [Google Scholar] [CrossRef]
  128. Chakravarty, D.; Ratnani, P.; Sobotka, S.; Lundon, D.; Wiklund, P.; Nair, S.S.; Tewari, A.K. Increased hospitalization and mortality from COVID-19 in prostate cancer patients. Cancers 2021, 13, 1630. [Google Scholar] [CrossRef]
  129. Drucker, D.J. Diabetes, obesity, metabolism, and SARS-CoV-2 infection: The end of the beginning. Cell Metab. 2021, 33, 479–498. [Google Scholar] [CrossRef]
  130. Steinke, I.; Ghanei, N.; Govindarajulu, M.; Yoo, S.; Zhong, J.; Amin, R.H. Drug discovery and development of novel therapeutics for inhibiting TMAO in models of atherosclerosis and diabetes. Front. Physiol. 2020, 11, 567899. [Google Scholar] [CrossRef]
  131. Constantino-Jonapa, L.A.; Espinoza-Palacios, Y.; Escalona-Montaño, A.R.; Hernández-Ruiz, P.; Amezcua-Guerra, L.M.; Amedei, A.; Aguirre-García, M.M. Contribution of trimethylamine N-oxide (TMAO) to chronic inflammatory and degenerative diseases. Biomedicines 2023, 11, 431. [Google Scholar] [CrossRef]
  132. Liu, M.-H.; Lin, X.-L.; Xiao, L.-L. SARS-CoV-2 nucleocapsid protein promotes TMAO-induced NLRP3 inflammasome activation by SCAP–SREBP signaling pathway. Tissue Cell 2024, 86, 102276. [Google Scholar] [CrossRef]
  133. Liu, M.-H.; Lin, X.-L.; Xiao, L.-L. Excess phosphate promotes SARS-CoV-2 N protein-induced NLRP3 inflammasome activation via the SCAP-SREBP2 signaling pathway. Mol. Med. Rep. 2024, 29, 1–12. [Google Scholar] [CrossRef]
  134. Strong, M.J.; McLellan, C.; Kaplanis, B.; Droppelmann, C.A.; Junop, M. Phase Separation of SARS-CoV-2 Nucleocapsid Protein with TDP-43 Is Dependent on C-Terminus Domains. Int. J. Mol. Sci. 2024, 25, 8779. [Google Scholar] [CrossRef]
  135. Kaji, S.; Maki, T.; Ishimoto, T.; Yamakado, H.; Takahashi, R. Insights into the pathogenesis of multiple system atrophy: Focus on glial cytoplasmic inclusions. Transl. Neurodegener. 2020, 9, 7. [Google Scholar] [CrossRef]
  136. Chen, X.; Jiang, B.; Gu, Y.; Yue, Z.; Liu, Y.; Lei, Z.; Yang, G.; Deng, M.; Zhang, X.; Luo, Z. SARS-CoV-2 nucleocapsid protein interaction with YBX1 displays oncolytic properties through PKM mRNA destabilization. Mol. Cancer 2024, 23, 248. [Google Scholar] [CrossRef]
  137. El-Maradny, Y.A.; Badawy, M.A.; Mohamed, K.I.; Ragab, R.F.; Moharm, H.M.; Abdallah, N.A.; Elgammal, E.M.; Rubio-Casillas, A.; Uversky, V.N.; Redwan, E.M. Unraveling the role of the nucleocapsid protein in SARS-CoV-2 pathogenesis: From viral life cycle to vaccine development. Int. J. Biol. Macromol. 2024, 279, 135201. [Google Scholar] [CrossRef]
  138. Mendoza-Ramírez, N.J.; García-Cordero, J.; Shrivastava, G.; Cedillo-Barrón, L. The key to increase immunogenicity of next-generation COVID-19 vaccines lies in the inclusion of the SARS-CoV-2 nucleocapsid protein. J. Immunol. Res. 2024, 2024, 9313267. [Google Scholar] [CrossRef]
  139. Kang, S.; Yang, M.; He, S.; Wang, Y.; Chen, X.; Chen, Y.Q.; Hong, Z.S.; Liu, J.; Jiang, G.M.; Chen, Q.Y.; et al. A SARS-CoV-2 antibody curbs viral nucleocapsid protein-induced complement hyperactivation. Nat. Commun. 2021, 12, 2697. [Google Scholar] [CrossRef]
  140. Dangi, T.; Sanchez, S.; Class, J.; Richner, M.; Visvabharathy, L.; Chung, Y.R.; Bentley, K.; Staton, R.J.; Koralnik, I.J.; Richner, J.M.; et al. Improved control of SARS-CoV-2 by treatment with a nucleocapsid-specific monoclonal antibody. J. Clin. Investig. 2022, 132, e162282. [Google Scholar] [CrossRef]
Figure 1. Proposed innate immune processes involving the N protein in host cells. Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are activated upon sensing dsRNA in the cytoplasm of infected cells. RIG-I and MDA5 initiate the mitochondrial antiviral signaling (MAVS) protein, thereby triggering a cascade of immune responses such as nuclear factor kappa B (NF-κB), interferon regulatory factor (IRF) 3, IRF7, and type I interferon (IFN-1) pathways. (a) SARS-CoV-2 N protein interacts with the RIG-I protein through the enzymatic active DExD/H domain, repressing the inflammatory response. (b) SARS-CoV-2 N protein interplayed with the TRIM25 and blocked RIG-I-mediated IFN production. (c) It also inhibited IFN signaling by targeting tank-binding kinase 1 (TBK1) and prevented the nuclear translocation of IRF3. Stress granules (SGs) are formed in response to viral infection as a host antiviral strategy. (d) SARS-CoV-2 N protein suppresses antiviral SG formation by interacting with GTPase-activating protein-binding protein 1 (G3BP1). (e) SARS-CoV-2 N protein undergoes LLPS with RNA during viral replication. (f) The formation of LLPS also inhibits SG assembly. (g) SARS-CoV-2 N protein also induces the expression of histone deacetylases 6 (HDAC6) to facilitate the interaction with G3BP1, thereby inhibiting the formation of SGs. (h) By sequestering growth arrest and DNA damage-inducible 34 (GADD34) mRNA into the N+foci and inhibiting its interaction with G3BP1, SARS-CoV-2 N protein further suppresses the transcription of IFN genes. (i) The SARS-CoV-2 N protein also suppressed the innate antiviral immune response by inhibiting Lys63-linked polyubiquitination and aggregation of MAVS. (j) SARS-CoV-2 N protein modulates MAVS SUMOylation by enhancing the interaction between UBC9 and MAVS. (k) SARS-CoV-2 N protein furthers the process of NF-κB signaling response and mitogen-activated protein kinase (MAPK) signaling pathway through Toll-like receptor 2 (TLR2). (l) During the LLPS process, N protein recruits key kinases of NF-κB signaling, such as TAK1 and IKK complex, increasing the possibility of the NF-κB activation. (m) The SARS-CoV-2 N protein binds to the receptor for advanced glycation end products (RAGE) via its NTD and CTD, activating the extracellular signal-regulated kinases 1 and 2 (ERK1/2)–NF-κB pathway. Nucleotide oligomerization domain-like receptor family pyrin domain containing-3 (NLRP3) functions in both inflammatory and antiviral responses. (n) SARS-CoV-2 infection promotes the activation of the NLRP3 inflammasome and caspase-1, (o) while GSDMD cleavage and pyroptosis are suppressed. RNA interference (RNAi) is a post-transcriptional gene silencing process. (p) SARS-CoV-2 N protein suppressed the RNAi in both initiation and effector steps by preventing siRNA biogenesis, RISC assembly, and target RNA cleavage, thereby evading the host inflammatory response. (q) Nonsense-mediated mRNA decay (NMD) is another antiviral mechanism involved in promoting mRNA regulation and degradation to prevent viral mRNAs translation. The SARS-CoV-2 N protein counteracts this mechanism by directly interacting with RNA helicase UP-frameshift-1 (UPF1) and UPF2. (r)The cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-dependent Cl channel, regulates the host immune defense against pathogen infection. SARS-CoV-2 N protein raises the intracellular Cl concentration by downregulating CFTR expression and depleting intracellular cAMP by increasing the phosphodiesterase 4 (PDE4), and the high intracellular Cl concentration induces the phosphorylation of serum glucocorticoid regulated kinase 1 (SGK1) and activates robust inflammatory responses. (s) SARS-CoV-2 N protein is ISGylated by HERC5 ISGylation.
Figure 1. Proposed innate immune processes involving the N protein in host cells. Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are activated upon sensing dsRNA in the cytoplasm of infected cells. RIG-I and MDA5 initiate the mitochondrial antiviral signaling (MAVS) protein, thereby triggering a cascade of immune responses such as nuclear factor kappa B (NF-κB), interferon regulatory factor (IRF) 3, IRF7, and type I interferon (IFN-1) pathways. (a) SARS-CoV-2 N protein interacts with the RIG-I protein through the enzymatic active DExD/H domain, repressing the inflammatory response. (b) SARS-CoV-2 N protein interplayed with the TRIM25 and blocked RIG-I-mediated IFN production. (c) It also inhibited IFN signaling by targeting tank-binding kinase 1 (TBK1) and prevented the nuclear translocation of IRF3. Stress granules (SGs) are formed in response to viral infection as a host antiviral strategy. (d) SARS-CoV-2 N protein suppresses antiviral SG formation by interacting with GTPase-activating protein-binding protein 1 (G3BP1). (e) SARS-CoV-2 N protein undergoes LLPS with RNA during viral replication. (f) The formation of LLPS also inhibits SG assembly. (g) SARS-CoV-2 N protein also induces the expression of histone deacetylases 6 (HDAC6) to facilitate the interaction with G3BP1, thereby inhibiting the formation of SGs. (h) By sequestering growth arrest and DNA damage-inducible 34 (GADD34) mRNA into the N+foci and inhibiting its interaction with G3BP1, SARS-CoV-2 N protein further suppresses the transcription of IFN genes. (i) The SARS-CoV-2 N protein also suppressed the innate antiviral immune response by inhibiting Lys63-linked polyubiquitination and aggregation of MAVS. (j) SARS-CoV-2 N protein modulates MAVS SUMOylation by enhancing the interaction between UBC9 and MAVS. (k) SARS-CoV-2 N protein furthers the process of NF-κB signaling response and mitogen-activated protein kinase (MAPK) signaling pathway through Toll-like receptor 2 (TLR2). (l) During the LLPS process, N protein recruits key kinases of NF-κB signaling, such as TAK1 and IKK complex, increasing the possibility of the NF-κB activation. (m) The SARS-CoV-2 N protein binds to the receptor for advanced glycation end products (RAGE) via its NTD and CTD, activating the extracellular signal-regulated kinases 1 and 2 (ERK1/2)–NF-κB pathway. Nucleotide oligomerization domain-like receptor family pyrin domain containing-3 (NLRP3) functions in both inflammatory and antiviral responses. (n) SARS-CoV-2 infection promotes the activation of the NLRP3 inflammasome and caspase-1, (o) while GSDMD cleavage and pyroptosis are suppressed. RNA interference (RNAi) is a post-transcriptional gene silencing process. (p) SARS-CoV-2 N protein suppressed the RNAi in both initiation and effector steps by preventing siRNA biogenesis, RISC assembly, and target RNA cleavage, thereby evading the host inflammatory response. (q) Nonsense-mediated mRNA decay (NMD) is another antiviral mechanism involved in promoting mRNA regulation and degradation to prevent viral mRNAs translation. The SARS-CoV-2 N protein counteracts this mechanism by directly interacting with RNA helicase UP-frameshift-1 (UPF1) and UPF2. (r)The cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-dependent Cl channel, regulates the host immune defense against pathogen infection. SARS-CoV-2 N protein raises the intracellular Cl concentration by downregulating CFTR expression and depleting intracellular cAMP by increasing the phosphodiesterase 4 (PDE4), and the high intracellular Cl concentration induces the phosphorylation of serum glucocorticoid regulated kinase 1 (SGK1) and activates robust inflammatory responses. (s) SARS-CoV-2 N protein is ISGylated by HERC5 ISGylation.
Viruses 17 01046 g001
Figure 2. N protein-mediated systemic inflammation.
Figure 2. N protein-mediated systemic inflammation.
Viruses 17 01046 g002
Figure 3. The interaction between long COVID and SARS-CoV-2 N protein.
Figure 3. The interaction between long COVID and SARS-CoV-2 N protein.
Viruses 17 01046 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cao, Y.; Wang, Y.; Huang, D.; Tan, Y.-J. The Role of SARS-CoV-2 Nucleocapsid Protein in Host Inflammation. Viruses 2025, 17, 1046. https://doi.org/10.3390/v17081046

AMA Style

Cao Y, Wang Y, Huang D, Tan Y-J. The Role of SARS-CoV-2 Nucleocapsid Protein in Host Inflammation. Viruses. 2025; 17(8):1046. https://doi.org/10.3390/v17081046

Chicago/Turabian Style

Cao, Yujia, Yaju Wang, Dejian Huang, and Yee-Joo Tan. 2025. "The Role of SARS-CoV-2 Nucleocapsid Protein in Host Inflammation" Viruses 17, no. 8: 1046. https://doi.org/10.3390/v17081046

APA Style

Cao, Y., Wang, Y., Huang, D., & Tan, Y.-J. (2025). The Role of SARS-CoV-2 Nucleocapsid Protein in Host Inflammation. Viruses, 17(8), 1046. https://doi.org/10.3390/v17081046

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop