COVID-19 and immune dysregulation: Genomic pathways, clinical manifestations, and therapeutic strategies

John Murphy, CEO & President, COVID-19 Long-haul Foundation

Abstract

SARS-CoV-2 provokes a uniquely heterogeneous and dynamic immune response that ranges from efficient viral control to fulminant hyperinflammation and prolonged immune dysregulation. Early viral sensing cascades, type I/III interferon programs, and downstream transcriptional circuits (NF-κB, JAK–STAT) determine whether the host transitions toward resolution or a pathogenic state characterized by lymphocyte dysregulation, myeloid overactivation, complement and coagulation pathway coupling, neutrophil extracellular trap (NET) formation, and inflammasome activation. Genetic variability in innate sensing and interferon signaling (e.g., OAS1, IFNAR2, TYK2), antigen presentation (HLA haplotypes), and inflammatory amplifiers (e.g., IL6/STAT3 axis) shape disease severity and the risk of persistent symptoms. This article synthesizes current understanding of COVID-19–associated immune malfunction, detailing the etiologic drivers and genomic architecture, the clinical and laboratory approach to diagnosis, and a pragmatic, evidence-based standard of care. We also survey emerging therapeutics—from interferon pathway rescue and inflammasome inhibition to complement/NET modulation and JAK/BTK targeting—that aim to suppress maladaptive immune responses in acute disease and post-acute sequelae.

Introduction

COVID-19 unfolded as an immunologic paradox: a virus often contained effectively by coordinated innate and adaptive responses but capable, in a subset, of uncoupling antiviral defense from inflammatory control. Severe disease typically follows a biphasic pattern—initially virus-dominant, later inflammation-dominant—where the second phase may be only partly virus-dependent and instead driven by host immune dysregulation. Across this trajectory, specific immune networks and genomic susceptibilities influence outcomes: type I/III interferon insufficiency; exuberant NF-κB–driven cytokine programs; monocyte, macrophage, and neutrophil hyperactivation; endothelial and complement activation; and, in some, a trajectory toward autoantibody production and long-term immune imbalance. Understanding these mechanisms is essential for precise clinical evaluation and for deploying stage-appropriate immunomodulatory therapy that blunts pathology without undermining needed antiviral control.

Etiology: How SARS-CoV-2 disrupts immune homeostasis

Early sensing and the interferon axis

  • Pattern recognition receptors (PRRs)—including TLR3/7, RIG-I (DDX58), and MDA5 (IFIH1)—detect SARS-CoV-2 RNA and initiate IRF3/7- and NF-κB–dependent transcription of type I/III interferons and proinflammatory mediators. In many patients with severe disease, these interferon programs are delayed, dampened, or antagonized by viral proteins (e.g., ORF6, NSP1) that block sensing, transcription, or STAT1 nuclear translocation. Insufficient early interferon signaling allows high viral replication and contiguous tissue damage, priming later hyperinflammation.
  • Inborn errors or functional antagonism of type I IFN (including neutralizing anti-IFN autoantibodies) are associated with severe COVID-19. Conversely, brisk early IFN responses correlate with milder disease, underscoring the timing-sensitive nature of IFN therapy.

Myeloid activation, cytokine circuits, and NF-κB/JAK–STAT integration

  • When viral control falters, myeloid cells amplify inflammation through NF-κB and JAK–STAT signaling, producing IL-6, TNF, IL-1β, GM-CSF, and chemokines (e.g., CCL2, CXCL8). Pathologic monocyte/macrophage states (HLA-DRlow, S100A8/A9high) expand, while emergency granulopoiesis yields immature and hyperreactive neutrophil subsets.
  • IL-6/STAT3 signaling drives acute-phase responses and endothelial activation; TNF and IL-1β reinforce tissue injury. Simultaneously, persistent antigenemia and cytokinemia push T cells toward exhaustion phenotypes (PD-1+, TIM-3+), reduce cytotoxic functionality, and skew CD4+ differentiation toward Th17 states, while regulatory T cell (Treg) function is often impaired.

Endothelium, complement, and coagulation coupling

  • SARS-CoV-2–infected or cytokine-primed endothelium expresses adhesion molecules and procoagulant tissue factor, supporting leukocyte recruitment and microthrombosis. Complement activation (lectin and alternative pathways) generates C3a/C5a anaphylatoxins that recruit and activate myeloid cells, while C5b-9 damages endothelium. This immunothrombotic loop is a hallmark of severe disease and is measurable through D-dimer elevation, fibrin deposition, and microvascular injury.

NETosis and inflammasome activation

  • Neutrophil extracellular traps (NETs)—DNA–histone webs decorated with proteases—trap virions but also amplify vascular injury and thrombosis. Simultaneously, NLRP3 inflammasome activation promotes IL-1β and IL-18 maturation, augments pyroptosis, and sustains sterile inflammation even as viral burden wanes.

Autoimmunity and post-acute immune dysregulation

  • Autoantibodies against phospholipids, cytokines (e.g., IFNs), and diverse self-antigens have been described during and after acute illness. Molecular mimicry, epitope spreading from tissue damage, bystander activation, and persistent antigenic stimulation (e.g., viral RNA/proteins retained in tissues) may collectively drive post-acute sequelae. Multi-omic studies reveal persistent innate activation, aberrant B cell maturation, and skewed TCR/BCR repertoires in some patients months after infection.

Genomic pathways and host susceptibility

Interferon signaling and antiviral restriction

  • TYK2, IFNAR2, OAS1, and genes in the 3p21.31 locus (including CCR2, CXCR6) are repeatedly implicated in severe disease. The OAS1 splice variant influenced by an introgressed Neanderthal haplotype enhances RNAse L–mediated degradation of viral RNA, conferring protection; reduced OAS1 activity correlates with worse outcomes. Reduced IFNAR2 expression or signaling impairs responsiveness to type I IFN, while TYK2 variants modify downstream STAT activation and inflammatory skew.
  • Inborn errors in TLR3, IRF7, and related genes, as well as neutralizing anti–type I IFN autoantibodies, have been causally linked to life-threatening COVID-19, highlighting the primacy of interferon biology in early control.

Antigen presentation and T cell coordination

  • HLA class I and II polymorphisms affect epitope presentation breadth and CD8+/CD4+ responses. Certain HLA supertypes present conserved SARS-CoV-2 epitopes more efficiently, sustaining cytotoxic T cell control despite viral evolution; other haplotypes associate with impaired presentation or immunopathology.

Chemokine trafficking and myeloid bias

  • Genetic signals at CCR2/CCR3 and CXCR6 influence leukocyte recruitment to the lung and inflamed tissues, shaping the balance between protective clearance and damaging infiltration.

Inflammasome and innate amplification

  • Polymorphisms in NLRP3, CARD8, and IL1B can potentiate inflammasome activity and cytokine release, fueling lung and systemic pathology.

Coagulation and endothelial regulation

  • Variants in ABO, F5, and PAI-1 (SERPINE1), while not COVID-specific, modulate thrombosis risk. Complement pathway genes (CFH, C3) may modify susceptibility to microangiopathy.

Epigenetic and transcriptomic reprogramming

  • Acute SARS-CoV-2 infection induces durable chromatin remodeling in innate cells (“trained immunity”) that can amplify responses to secondary stimuli. DNA methylation signatures in interferon-stimulated genes and exhaustion markers persist in some individuals, correlating with symptom burden in post-acute sequelae.

Clinical picture and physical diagnosis of immune dysregulation

Acute immune pathology

  • Patients with severe disease often present with fever, tachypnea, hypoxemia, and evidence of systemic inflammation (marked fatigue, myalgias). On examination, signs of respiratory distress dominate, but clues to immune dysregulation include livedo reticularis or acro-ischemia (microthrombi), neurologic fluctuations (encephalopathy), conjunctivitis, and myopericardial involvement. A hyperinflammatory state may evolve around days 7–12 from symptom onset, when viral replication wanes but host inflammation escalates.

Post-acute sequelae (PASC) patterns

  • Weeks to months after infection, some patients show orthostatic intolerance (POTS-like features), cognitive deficits (“brain fog”), exertional intolerance, chest pain, dyspnea, and musculoskeletal pain. Physical exam may reveal tachycardia on standing, distal dysesthesias, tender points, and variable joint swelling or enthesitis. These findings, while nonspecific, prompt a laboratory and imaging strategy to phenotype immune activity and rule out alternative etiologies.

Laboratory and imaging evaluation

Core inflammatory and coagulation panel

  • CBC with differential: lymphopenia and neutrophilia correlate with severity; immature granulocytes and low absolute lymphocyte count suggest myeloid-skewed inflammation.
  • CRP and ferritin: track IL-6–driven acute-phase responses; extreme ferritin raises concern for macrophage activation.
  • D-dimer, fibrinogen, PT/aPTT: assess immunothrombotic activity; elevated D-dimer aligns with microvascular injury.
  • LDH and transaminases: reflect tissue injury.

Cytokines and immune cell phenotyping (as available)

  • IL-6, IL-1β, TNF, IL-10, IFN-α/β activity or transcriptional “IFN signature” (e.g., ISG expression) can stratify phases; high IL-6 with muted IFN suggests late hyperinflammation.
  • Flow cytometry: CD4/CD8 counts; activation/exhaustion markers (HLA-DR, CD38, PD-1, TIM-3); Treg frequencies (CD25+FOXP3+); memory B cell and plasmablast fractions. Elevated low-density neutrophils and HLA-DRlow monocytes indicate maladaptive myeloid activation.

Autoimmunity, complement, and NETs

  • ANA, ENA panel, rheumatoid factor, anti-CCP, antiphospholipid antibodies (aCL, anti-β2GPI) and anti-cytokine antibodies (if accessible) characterize autoimmune features.
  • Complement (C3, C4), CH50/AH50, and sC5b-9 reflect complement consumption/activation.
  • Cell-free DNA, MPO–DNA complexes, and citrullinated histone H3 provide NETosis readouts in specialized centers.

Virology and antigenemia

  • Quantitative PCR with cycle thresholds (trend), antigenemia assays, and, in select cases, tissue sampling for viral RNA/protein detection can clarify whether ongoing antigen drives inflammation.

Imaging

  • Chest CT: organizing pneumonia, microvascular patterns; V/Q scans or CTPA for thromboembolism.
  • Echocardiography and cardiac MRI for myocarditis; neuroimaging for encephalopathy; vascular ultrasound for thrombosis.

Standard of care: Stage-appropriate treatment

Care must match disease phase: promote viral clearance early; dampen maladaptive inflammation when host injury dominates; maintain vigilance for thrombosis and organ-specific complications.

Antivirals

  • Early outpatient therapy within the viral-replication window reduces progression and downstream inflammation: nirmatrelvir–ritonavir, 3-day remdesivir, and selected monoclonal antibodies when variant-active. In hospitalized patients with ongoing replication, remdesivir shortens time to recovery.

Corticosteroids

  • Dexamethasone (or equivalent) reduces mortality in patients requiring supplemental oxygen or mechanical ventilation by curbing NF-κB–mediated cytokine cascades, decreasing endothelial activation, and restoring lymphocyte homeostasis. Avoid routine steroids in patients not requiring oxygen, where antiviral control remains the priority.

IL-6 pathway inhibition

  • Tocilizumab or sarilumab, when layered on corticosteroids in rapidly progressive, systemically inflamed, oxygen-requiring patients, reduces progression to ventilation and death. Selection is guided by escalating oxygen needs, CRP elevation, and absence of uncontrolled bacterial infection.

JAK inhibition

  • Baricitinib (and, in some settings, tofacitinib) attenuates JAK–STAT signaling downstream of multiple cytokines (IL-6, IFN-γ, GM-CSF), reducing time to recovery and mortality in hypoxemic patients—particularly when added to remdesivir and/or steroids.

IL-1 inhibition and hyperinflammation

  • Anakinra, guided by biomarkers of macrophage activation (e.g., ferritin, suPAR), can be beneficial in select hyperinflammatory phenotypes, reflecting a role for inflammasome-mediated IL-1 signaling.

Anticoagulation

  • Prophylactic-dose anticoagulation is standard for hospitalized patients; in non-ICU patients with high D-dimer and low bleeding risk, therapeutic-dose heparin can reduce the need for organ support. Post-discharge extended prophylaxis is individualized.

MIS-C and MIS-A paradigms

  • Pediatric multisystem inflammatory syndrome (MIS-C) and adult analogs (MIS-A) often respond to IVIG and corticosteroids; escalation to anakinra or infliximab is considered in refractory cases. Cardiac monitoring is essential.

Supportive and organ-directed care

  • Oxygenation strategies (HFNC, prone positioning), lung-protective ventilation, renal replacement therapy as needed, and treatment of secondary infections remain core. Nutritional and rehabilitation support preserve immune and functional recovery capacity.

Managing post-acute immune dysregulation (PASC)

PASC care requires phenotype-driven pragmatism and careful exclusion of alternative causes.

  • Autonomic dysfunction: graded fluid/salt loading, compression garments, low-dose beta blockers or ivabradine, and autonomic rehabilitation can stabilize POTS-like symptoms.
  • Mast cell–like syndromes: H1/H2 antihistamines, leukotriene antagonists, cromolyn (where available), and trigger avoidance may alleviate flushing, urticaria, and GI dysmotility in a subset.
  • Inflammatory/musculoskeletal symptoms: paced activity to avoid post-exertional exacerbation, targeted physical therapy, neuropathic pain agents, and, in documented inflammatory arthritis or myositis, conventional DMARDs under rheumatologic guidance.
  • Sleep and mood: address sleep fragmentation, treat mood disorders, and incorporate cognitive rehabilitation strategies for executive dysfunction.
  • Investigational and specialty-guided care: off-label low-dose naltrexone shows promise in reducing neuroinflammation and pain in observational contexts; immunologic trials (e.g., JAK inhibition, complement modulation, antivirals for suspected viral persistence) are underway but should be pursued within research frameworks whenever possible.

Therapeutic research frontiers

Restoring antiviral signaling

  • Type I/III interferon therapy is timing-sensitive; inhaled or early parenteral administration is under renewed study for high-risk outpatients. Downstream rescue via TYK2/JAK1-selective modulation aims to balance antiviral tone without precipitating hyperinflammation.

Inflammasome and IL-1 axis

  • Small-molecule NLRP3 inhibitors (e.g., dapansutrile) and IL-1 blockade strategies are being tested to tame macrophage-driven injury, particularly in patients with inflammasome gene signatures.

Complement and endothelium

  • C5 or C5a blockade and lectin pathway inhibition (e.g., anti-MASP-2) target the immunothrombotic loop fueling organ damage. Endothelial stabilizers and antiplatelet strategies are being evaluated in precise phenotypes.

NETosis modulation

  • DNase-based NET dismantling and peptidylarginine deiminase (PAD) inhibition represent mechanistic approaches to reduce microvascular injury; optimal patient selection and timing remain active areas of investigation.

BTK and GM-CSF targeting

  • Bruton’s tyrosine kinase inhibitors modulate FcγR and TLR signaling in myeloid cells; anti–GM-CSF antibodies aim to interrupt monocyte–macrophage amplification pathways.

B cell and autoantibody strategies

  • For select autoantibody-mediated complications, B cell depletion or plasma exchange has mechanistic rationale; prospective trials will define risk–benefit in post-acute disease.

Multi-omic personalization

  • Longitudinal immune “endotyping” (cell states, cytokine circuits, TCR/BCR clonality, metabolomics) is guiding adaptive platform trials to match immunomodulators to the dominant biology in a given patient and timepoint.

Practical clinical framework

  1. Classify the phase and dominant biology:
    • Early viral replication vs. hyperinflammation vs. mixed; acute vs. post-acute.
  2. Establish severity and thrombotic risk:
    • Oxygen needs, organ involvement, D-dimer, imaging.
  3. Apply stage-appropriate therapy:
    • Early antivirals; oxygenation; when hyperinflammation emerges, add corticosteroids and consider IL-6 or JAK inhibitors; integrate anticoagulation thoughtfully.
  4. Phenotype immunopathology in complex or persistent cases:
    • Cytokines, autoantibodies, complement, cell phenotypes, and imaging to guide targeted interventions.
  5. Monitor and adjust:
    • Track CRP, ferritin, lymphocyte counts, oxygenation, and organ function; taper immunomodulation as biology resolves.
  6. Plan recovery:
    • Rehabilitation, autonomic management, sleep/mood care; revisit immune phenotyping if plateaus or relapses occur.

Conclusion

COVID-19 immune dysregulation is not a single entity but a set of shifting immunologic states determined by the kinetics of viral control, host genomic wiring, and the resilience—or fragility—of core regulatory networks. Interferon competence, balanced myeloid activation, intact endothelial and complement homeostasis, and preserved lymphocyte function are the pillars of favorable outcomes. Standard-of-care therapy succeeds when it aligns with these principles and the patient’s phase-specific biology: antivirals early; judicious immunomodulation once host injury dominates; anticoagulation to mitigate immunothrombosis. The next advances will come from precise endotyping and targeted interventions that prevent the immune system from tipping into self-damage—during acute infection and across the long arc of recovery.

References

  1. Blanco-Melo D, Nilsson-Payant BE, Liu WC, et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell. 2020;181(5):1036–1045.e9.
  2. Hadjadj J, Yatim N, Barnabei L, et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science. 2020;369(6504):718–724.
  3. Zhang Q, Bastard P, Liu Z, et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science. 2020;370(6515):eabd4570.
  4. Bastard P, Rosen LB, Zhang Q, et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020;370(6515):eabd4585.
  5. Pairo-Castineira E, Clohisey S, Klaric L, et al. Genetic mechanisms of critical illness in COVID-19. Nature. 2021;591(7848):92–98.
  6. Ellinghaus D, Degenhardt F, Bujanda L, et al. Genomewide association study of severe COVID-19 with respiratory failure. N Engl J Med. 2020;383(16):1522–1534.
  7. Huffman JE, Butler-Laporte G, Khan A, et al. Multi-ancestry fine-mapping implicates OAS1 splice variant in protection against COVID-19 severity. Nat Med. 2021;27(11):1793–1805.
  8. Mathew D, Giles JR, Baxter AE, et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science. 2020;369(6508):eabc8511.
  9. Silvin A, Chapuis N, Dunsmore G, et al. Elevated calprotectin and abnormal myeloid cell subsets discriminate severe COVID-19 patients. Nat Immunol. 2020;21(8):982–987.
  10. Lucas C, Wong P, Klein J, et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature. 2020;584(7821):463–469.
  11. Woodruff MC, Ramonell RP, Nguyen DC, et al. Extrafollicular B cell responses correlate with neutralizing antibodies and morbidity in COVID-19. Nat Immunol. 2020;21(12):1506–1516.
  12. Zuo Y, Yalavarthi S, Shi H, et al. Neutrophil extracellular traps in COVID-19. JCI Insight. 2020;5(11):e138999.
  13. Magro C, Mulvey JJ, Berlin D, et al. Complement associated microvascular injury and thrombosis in severe COVID-19. Transl Res. 2020;220:1–13.
  14. Rodrigues TS, de Sá KSG, Ishimoto AY, et al. Inflammasomes are activated in response to SARS-CoV-2 infection and associated with COVID-19 severity in patients. J Exp Med. 2021;218(3):e20201707.
  15. Yang OO. The immunopathogenesis of SARS-CoV-2 infection: overview of lessons learned in the first 5 years. J Immunol. 2025;214(6):1095–1104.
  16. RECOVERY Collaborative Group. Dexamethasone in hospitalized patients with Covid-19. N Engl J Med. 2021;384(8):693–704.
  17. REMAP-CAP Investigators. Interleukin-6 receptor antagonists in critically ill patients with Covid-19. N Engl J Med. 2021;384(16):1491–1502.
  18. 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.
  19. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the treatment of Covid-19—final report. N Engl J Med. 2020;383(19):1813–1826.
  20. ATTACC, ACTIV-4a, and REMAP-CAP Investigators. Therapeutic anticoagulation with heparin in noncritically ill patients with Covid-19. N Engl J Med. 2021;385(9):790–802.
  21. Salama C, Han J, Yau L, et al. Tocilizumab in patients hospitalized with Covid-19 pneumonia. N Engl J Med. 2021;384(1):20–30.
  22. Kyriazopoulou E, Huet T, Cavalli G, et al. Effect of anakinra on mortality in patients with COVID-19 and hyperinflammation: a randomized clinical trial. Lancet Rheumatol. 2021;3(7):e457–e466.
  23. Su Y, Yuan D, Chen DG, et al. Multiple early factors anticipate post-acute COVID-19 sequelae. Nature. 2022;606(7916):430–439.
  24. Scully EP, Haverfield J, Ursin RL, et al. Consideration of sex as a biological variable in COVID-19. Nat Rev Immunol. 2020;20(7):442–447.
  25. Mehandru S, Merad M. Pathological sequelae of long-haul COVID. Nat Immunol. 2022;23(2):194–202.

Leave a Reply

Your email address will not be published. Required fields are marked *