DSG2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
ARVC 10 antibody; ARVC10 antibody; ARVD 10 antibody; ARVD10 antibody; Cadherin family member 5 antibody; CDHF 5 antibody; CDHF5 antibody; CMD1BB antibody; Desmoglein 2 antibody; Desmoglein-2 antibody; Desmoglein2 antibody; DSG 2 antibody; DSG2 antibody; DSG2_HUMAN antibody; HDGC antibody; HDGC included antibody; Human Desmoglein colon antibody; MGC117034 antibody; MGC117036 antibody; MGC117037 antibody
Target Names
Uniprot No.

Target Background

Function
Desmoglein-2 (DSG2) is a component of intercellular desmosome junctions. It plays a role in the interaction of plaque proteins and intermediate filaments, thereby mediating cell-cell adhesion.
Gene References Into Functions
  1. A homozygous mutation of DSG2 p.F531C was identified as the pathogenic mutation in patients with arrhythmogenic right ventricular cardiomyopathy/dysplasia (ARVC/D) involving both ventricles. This mutation leads to widened and impaired intercalated discs, interrupted myocardial fibers, and abnormally hyperplastic interstitial fibers, collagen fibers, and adipocytes. PMID: 28578331
  2. DSG2 is a key regulator of vasculogenic mimicry (VM) activity in human melanoma. This suggests that targeting DSG2 might be a potential therapeutic strategy to reduce tumor blood supply and metastatic spread. PMID: 27340778
  3. Data suggest that Dsg2 stimulates cell growth and migration by positively regulating EGFR level and signaling through a c-Src and Cav1-dependent mechanism utilizing lipid rafts as signal modulatory platforms. PMID: 26918609
  4. Studies have identified DSG2 expression in distinct progenitor cell subpopulations and demonstrate that, independent of its traditional function as a component of desmosomes, this cadherin also plays a critical role in the vasculature. PMID: 27338829
  5. Expression of the desmosomal protein Desmoglein-2 was found to be reduced in pediatric dilated cardiomyopathy patients. PMID: 28764973
  6. This study defines a mechanism by which Dsg2 expression in cancer cells can modulate the tumor microenvironment, a step crucial for tumor progression. PMID: 28438789
  7. Silencing of Dsg2, but not Dsc2, resulted in loss of cell cohesion and enhanced migration and invasion of pancreatic adenocarcinoma cells. PMID: 28277619
  8. The homozygous desmoglein 2 variant c.1003A;G co-segregated with Arrhythmogenic right ventricular cardiomyopathy, indicating autosomal recessive inheritance and complete penetrance. PMID: 28818065
  9. These data suggest that palmitoylation of Dsg2 regulates protein transport to the plasma membrane. Modulation of the palmitoylation status of desmosomal cadherins can affect desmosome dynamics. PMID: 27703000
  10. Both Dsg2 mRNA and protein were highly expressed in non-small cell lung cancer (NSCLC) tissues and associated with NSCLC size, but not with overall survival of patients. PMID: 27629878
  11. Currently, 13 genes have been associated with the disease but nearly 40% of clinically diagnosed cases remain without a genetic diagnosis. PMID: 25398255
  12. DSG2 and DSG3 might be potential diagnostic markers for squamous cell carcinoma of the lung. PMID: 25468811
  13. In endometrial luminal epithelium, cadherin 6, desmoglein 2, and plexin b2 were surprisingly found in the apical as well as the lateral membrane domain. Their knockdown compromised epithelial integrity. PMID: 25237006
  14. A low DSG2 expression phenotype is a useful prognostic biomarker of tumor aggressiveness and may serve as an aid in identifying patients with clinically significant prostate cancer. PMID: 24896103
  15. Six variants of uncertain clinical significance in the PKP2, JUP, and DSG2 genes showed a deleterious effect on mRNA splicing, indicating these are ARVD/C-related pathogenic splice site mutations. PMID: 25087486
  16. This structure reveals that the ectodomain of Dsg2 is flexible even in the calcium-bound state and, on average, is shorter than the type 1 cadherin crystal structures. PMID: 25855637
  17. Desmoglein 2 expression attenuates migration of melanoma cells, mediated by downregulation of secretogranin II. PMID: 24558503
  18. Gal3 has a role in stabilizing desmoglein-2, a desmosomal cadherin, and intercellular adhesion in intestinal epithelial cells. PMID: 24567334
  19. Desmoglein-2 co-localizes with integrin-beta8 in N-MVECs. PMID: 23874518
  20. Authors found a number of mutations within or near the EF loop of the Ad3 fiber knob that resulted in affinities to DSG2 that were several orders of magnitude higher than those to the wild-type Ad3 knob. PMID: 23946456
  21. Findings were consistent with the results obtained by immunohistochemistry of endomyocardial biopsies and epidermal tissue of mutation carriers, which indicated a normal cellular distribution of DSG2. PMID: 23381804
  22. Snail regulates levels of E-cadherin and desmoglein 2 in oral squamous cell carcinoma cells both transcriptionally and post-translationally. PMID: 23261431
  23. CD133 interacts with plakoglobin, and knockdown of CD133 by RNA interference (RNAi) results in the downregulation of desmoglein-2. PMID: 23326490
  24. Specific desmosomal cadherins contribute differently to keratinocyte cohesion, and Dsg2 compared to Dsg3 is less important in this context. PMID: 23326495
  25. An impaired prodomain cleavage and an influence on the DSG2 properties could be demonstrated for the R46Q-variant, leading to the classification of the variant as a potential gain-of-function mutant in arrhythmogenic right ventricular cardiomyopathy. PMID: 23071725
  26. The Dsg unique region (DUR) of Dsg2 stabilized Dsg2 at the cell surface by inhibiting its internalization and promoted strong intercellular adhesion. PMID: 23128240
  27. Gastroesophageal reflux disease was specifically associated with elevated transcript levels of desmoglein 2 and plakoglobin. PMID: 22521077
  28. Dsg-2 with a mutation at the predicted cleavage site is resistant to cleavage by matriptase. Thus, Dsg-2 seems to be a functionally relevant physiological substrate of matriptase. PMID: 22783993
  29. Desmoglein 2, expressed earliest among the four isoforms in development, was found to be mutated in arrythmogenic right ventricular cardiomyopathy and is a receptor for a subset of adenoviruses that cause respiratory and urinary tract infections. PMID: 22189787
  30. The Dsg2 exhibits microtubule-dependent transport in epithelial cells but use distinct motors to traffic to the plasma membrane. PMID: 22184201
  31. We detected a novel mutation: DSG2 3059_3062delAGAG, and it may induce disintegration of the desmosomal structure. PMID: 21397041
  32. Dsg2 extracellular and intracellular domains are cleaved by proteolytic enzymes, and multiple cleavage fragments of Dsg2 are generated in colonic epithelial cells. PMID: 21715983
  33. Study demonstrated a molecular switching in gene expression within the desmoglein subfamily between DSG3 and DSG2 during oral cancer progression. PMID: 20923451
  34. Co-segregation of the G812S mutation with disease expression was established in a large Caucasian family. No differences in targeting or stability of the mutant proteins, suggesting that they act via a dominant negative mechanism. PMID: 20708101
  35. Dsg2-mediated adhesion affects tight junction integrity and is required to maintain intestinal epithelial barrier properties. PMID: 20224006
  36. Desmoglein 2 was highly expressed by the least differentiated cells of the cutaneous epithelium, including the hair follicle bulge of the fetus and adult, bulb matrix cells, and basal layer of the outer root sheath. PMID: 12787134
  37. Nine heterozygous DSG2 mutations (5 missense, 2 insertion-deletions, 1 nonsense, and 1 splice site mutation) were detected in subjects with ARVC. PMID: 16505173
  38. Mutations in DSG2 contribute to the development of arrhythmogenic right ventricular dysplasia/cardiomyopathy. PMID: 16773573
  39. Data demonstrate that UV-induced desmoglein-2 down-regulation is mediated via reactive oxygen species which are generated through EGF receptor activation and Rac2/NADPH oxidase activation. PMID: 16820949
  40. Mutations in DSG2 display a high degree of penetrance. Disease expression was of variable severity with left ventricular involvement a prominent feature. PMID: 17105751
  41. Long-term treatment with epidermal growth factor (EGF) leads to a marked increase in the levels of ADAM17, which also increases the shedding of several substrates of ADAM17, including the desmosomal cadherin Dsg-2. PMID: 17227756
  42. Desmoglein 2 is a novel solitary surface glycoprotein in malignant melanoma cells. PMID: 17495963
  43. Dsg2 was targeted by caspases in cell lines undergoing staurosporine-induced apoptosis. The proteolytic processing of full-length Dsg2 released a 70-kDa fragment into the cytosol. PMID: 17559062
  44. Dsg2 regulates intestinal epithelial cell apoptosis driven by cysteine proteases during physiological differentiation and inflammation. PMID: 17804817
  45. DSG2-V55M polymorphism is identified as a novel risk variant for dilated cardiomyopathy. PMID: 18678517
  46. Monoclonal antibodies against the proregion of the desmosomal cadherin, human desmoglein-2. PMID: 18707543
  47. Desmoglein 2 has been demonstrated in a sizable subset of nevi and primary melanomas. PMID: 18975006
  48. Results show that epidermal growth factor receptor inhibition stabilizes desmoglein 2 at intercellular junctions by interfering with its accumulation in an internalized cytoplasmic pool. PMID: 18987342
  49. Levels of Dsg1 & Dsg2 are reduced in pancreatic tumors; expression of kallikrein 7 in BxPC-3 cells resulted in an increase in shedding of soluble Dsg2. PMID: 19091121
  50. While Dsg2 expression was consistently strong in BCC, it varied in SCC with a minor correlation between a decrease of Dsg2 expression and tumor differentiation. PMID: 19458482

Show More

Hide All

Database Links

HGNC: 3049

OMIM: 125671

KEGG: hsa:1829

STRING: 9606.ENSP00000261590

UniGene: Hs.412597

Involvement In Disease
Arrhythmogenic right ventricular dysplasia, familial, 10 (ARVD10); Cardiomyopathy, dilated 1BB (CMD1BB)
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Cell junction, desmosome.
Tissue Specificity
All of the tissues tested and carcinomas.

Q&A

What is DSG2 and what are the key biological functions of the protein that DSG2 antibodies target?

DSG2 (Desmoglein 2) is a 122.3 kDa protein comprising 1118 amino acids with membrane subcellular localization. It functions primarily as a cell adhesion molecule in desmosomes, which are specialized cell junctions providing mechanical strength to tissues. DSG2 plays crucial roles in calcium ion binding, apoptosis regulation, development, and signal transduction . As the largest desmoglein, it is expressed in all desmosome-containing tissues, highlighting its fundamental role in maintaining cellular architecture and tissue integrity . DSG2 undergoes post-translational modifications including palmitoylation and glycosylation . Anti-DSG2 antibodies specifically recognize and bind to this protein, enabling researchers to study its expression, localization, and function in various experimental contexts.

How do monoclonal and polyclonal DSG2 antibodies differ in research applications?

Monoclonal DSG2 antibodies:

  • Recognize a single epitope on the DSG2 protein

  • Provide high specificity and reduced background in immunoassays

  • Offer consistent lot-to-lot reproducibility

  • Ideal for applications requiring precise epitope targeting such as co-immunoprecipitation and epitope mapping

  • Examples include the mouse monoclonal AH12.2 antibody that detects DSG2 protein of human origin

Polyclonal DSG2 antibodies:

  • Recognize multiple epitopes on the DSG2 protein

  • Provide higher sensitivity due to multiple binding sites

  • More tolerant to minor protein denaturation or modification

  • Better for applications like Western blotting where protein may be partially denatured

  • Examples include polyclonal goat anti-human DSG2 antibodies used for immunoassay validation

The choice between these antibody types depends on the specific research requirements, with monoclonals preferred for precise epitope detection and polyclonals for enhanced signal detection across various applications.

What experimental techniques are commonly used with DSG2 antibodies?

DSG2 antibodies are versatile research tools employed across multiple experimental platforms:

ApplicationDescriptionConsiderations
Western BlottingDetection of DSG2 protein in cell/tissue lysatesEffective for quantitative analysis of expression levels
ImmunoprecipitationIsolation of DSG2 and associated proteinsUseful for studying protein-protein interactions
ImmunofluorescenceVisualization of DSG2 localization in cells/tissuesProvides spatial information about protein distribution
ImmunohistochemistryDetection of DSG2 in tissue sectionsReveals expression patterns in pathological specimens
ELISAQuantitative measurement of DSG2 or anti-DSG2 antibodiesSuitable for high-throughput screening
Flow CytometryAnalysis of DSG2 expression in cell populationsAllows for single-cell resolution analysis
Electrochemiluminescent ImmunoassaysDetection of anti-DSG2 autoantibodies in serumUsed in clinical research for autoantibody detection

When designing experiments, researchers should consider the appropriate antibody format (native or conjugated) based on their specific application needs. Conjugated forms include agarose, HRP, PE, FITC, and various Alexa Fluor® conjugates .

How are DSG2 antibodies used to study post-COVID-19 cardiac pathology?

Researchers have employed DSG2 antibodies to investigate the emerging link between COVID-19 infection and cardiac sequelae through several sophisticated approaches:

  • Development of specialized immunoassays: Electrochemiluminescent-based immunoassays utilizing the extracellular domain of DSG2 for antibody capture have been developed to detect anti-DSG2 autoantibodies in patient serum. These assays are validated using commercial anti-DSG2 antibodies as positive controls .

  • Longitudinal analysis of autoantibody persistence: Studies have tracked anti-DSG2 autoantibody levels in post-COVID-19 patients at multiple timepoints (6 and 9 months post-infection), revealing sustained elevation of these antibodies well into recovery .

  • Comparative analysis with control populations: Research has demonstrated that post-COVID-19 patients exhibit significantly higher mean signal intensity of anti-DSG2 antibodies compared to healthy control populations (19±83.2 vs. 2.1±6.8, P<0.001), with approximately 29% of patients showing levels above the 90th percentile of controls .

  • Correlation with ARVC pathology: Approximately 8.7% of post-COVID-19 patients demonstrated anti-DSG2 antibody levels higher than the median found in Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC) patients, suggesting potential mechanistic overlaps between post-COVID cardiac manifestations and established cardiomyopathies .

This research supports the hypothesis that SARS-CoV-2 infection may trigger autoimmunity by exposing previously hidden cryptic epitopes on damaged cardiac cells to an activated immune system, potentially explaining the high incidence of cardiac involvement in COVID-19 .

What is the role of anti-DSG2 antibodies in targeting pluripotent stem cells?

Anti-DSG2 antibodies have been engineered as innovative tools to address a critical challenge in stem cell therapy—the risk of teratoma formation due to residual undifferentiated cells. The methodology employs several sophisticated approaches:

  • Antibody-drug conjugate (ADC) development: Researchers have conjugated monoclonal antibodies against DSG2 (specifically the K6-1 antibody) with doxorubicin (DOX), a chemotherapeutic agent. This creates a targeted delivery system specific to cells expressing high levels of DSG2 .

  • Selective targeting mechanism: The approach exploits differential expression of DSG2, which is highly expressed in undifferentiated human pluripotent stem cells (hPSCs) but has minimal expression in differentiated somatic tissues. This expression pattern provides a biological basis for selective targeting .

  • Cellular processing pathway: Upon binding to DSG2 on hPSCs, the K6-1-DOX conjugates are internalized through receptor-mediated endocytosis. Inside the cell, the acidic pH of endosomes triggers the release of doxorubicin, which subsequently localizes to the nucleus and induces cytotoxicity via an apoptotic caspase cascade .

  • Specificity validation: In contrast to DSG2-positive hPSCs, DSG2-negative fibroblasts showed minimal conjugate uptake or cytotoxicity, confirming the specificity of the approach and suggesting limited off-target effects .

  • In vivo confirmation: Mouse xenograft models have validated this approach, showing that hPSCs pretreated with K6-1-DOX conjugates failed to form teratomas when injected, while those treated with control hIgG-DOX developed teratomas .

This methodology represents a significant advance in improving the safety profile of stem cell therapies by providing a means to selectively eliminate potentially tumorigenic undifferentiated cells while preserving the therapeutic differentiated cell populations.

How do anti-DSG2 autoantibodies contribute to cardiac pathogenesis in ARVC?

Anti-DSG2 autoantibodies have emerged as important factors in the pathogenesis of Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC), a condition characterized by fibrofatty replacement of cardiomyocytes. The pathogenic mechanisms involve:

  • Autoimmune targeting: Anti-DSG2 autoantibodies specifically target the desmosomal protein DSG2, which is essential for mechanical integrity of cardiac tissue. Recent research has demonstrated that these autoantibodies are specific for ARVC and absent in healthy subjects and patients with other forms of cardiac disease .

  • Disruption of desmosomal adhesion: When anti-DSG2 autoantibodies bind to DSG2, they can interfere with calcium-dependent homophilic binding between DSG2 molecules on adjacent cardiomyocytes, compromising desmosomal integrity.

  • Induction of signaling cascades: Binding of autoantibodies to DSG2 can trigger intracellular signaling pathways that promote apoptosis, fibrosis, and inflammatory responses in cardiac tissue.

  • Functional evidence: Studies have shown that anti-DSG2 antibodies from ARVC patients cause direct cardiac pathology in vitro, suggesting a functional role in disease progression rather than merely serving as disease markers .

  • Persistence and progression: The sustained presence of these autoantibodies contributes to ongoing tissue damage and disease progression, as evidenced by the persistent elevation of anti-DSG2 antibodies in longitudinal studies .

Understanding these mechanisms is crucial for developing targeted therapies for ARVC and potentially for post-COVID-19 cardiac sequelae, which show similar autoantibody patterns. This research highlights the broader importance of desmosomal autoimmunity in cardiac pathology.

What are the optimal methods for validating anti-DSG2 antibody specificity?

Rigorous validation of anti-DSG2 antibodies is essential for ensuring experimental reliability. A comprehensive validation strategy includes:

  • Western blot analysis:

    • Use multiple cell/tissue lysates with known DSG2 expression levels

    • Include positive controls (tissues with high DSG2 expression like cardiac tissue)

    • Include negative controls (DSG2 knockout cells/tissues)

    • Verify that the detected band corresponds to the expected molecular weight (122.3 kDa)

  • Immunoprecipitation validation:

    • Perform reciprocal IP-Western experiments using different anti-DSG2 antibodies

    • Confirm specificity using mass spectrometry analysis of immunoprecipitated proteins

    • Assess cross-reactivity with other desmoglein family members (DSG1, DSG3, DSG4)

  • Immunofluorescence/immunohistochemistry controls:

    • Compare staining patterns with established desmosomal markers

    • Perform peptide competition assays to confirm epitope specificity

    • Use siRNA knockdown or knockout models to demonstrate staining specificity

  • Cross-species reactivity testing:

    • Test antibody against DSG2 from multiple species if cross-reactivity is claimed

    • Human DSG2 should be the primary validation target, with testing for orthologues in mouse, rat, bovine, frog, chimpanzee, and chicken as needed

  • Application-specific validation:

    • For specialized applications like electrochemiluminescent immunoassays, validate using commercially available anti-DSG2 antibodies as reference standards

    • Document detection limits and dynamic range for quantitative applications

Proper documentation of validation experiments is essential for reproducibility and should be included in publications and experimental protocols.

How should researchers design experiments to study anti-DSG2 autoantibodies in patient samples?

When investigating anti-DSG2 autoantibodies in clinical research, several methodological considerations are crucial:

  • Immunoassay development and optimization:

    • Utilize recombinant extracellular domain of DSG2 as the capture antigen for autoantibody detection

    • Validate assay performance using commercial anti-DSG2 antibodies as positive controls

    • Establish standardized signal normalization protocols using negative control samples

  • Study cohort design:

    • Include appropriate control groups (healthy subjects, disease controls with other cardiac conditions)

    • Match cohorts for demographic factors (age, sex, ethnicity)

    • For post-infectious studies, clearly define timepoints relative to infection (e.g., 6 and 9 months post-COVID-19)

  • Longitudinal sampling strategy:

    • Collect paired samples from the same patients at multiple timepoints to assess persistence and dynamics of autoantibody responses

    • Standardize sample collection, processing, and storage procedures

  • Statistical analysis approach:

    • Define cut-off values based on percentiles (e.g., 90th percentile) of healthy control populations

    • Use appropriate statistical tests for comparing autoantibody levels between groups

    • Account for multiple comparisons when analyzing various timepoints or subgroups

  • Correlative analyses:

    • Integrate clinical data on cardiac function/symptoms with autoantibody measurements

    • Consider multi-parameter analysis combining different autoantibody specificities

    • Correlate autoantibody levels with disease severity markers or outcomes

By following these guidelines, researchers can generate robust data on anti-DSG2 autoantibodies that contributes meaningfully to understanding their role in cardiac pathology following infections or in cardiomyopathies.

What technical challenges exist in developing antibody-drug conjugates targeting DSG2, and how can they be addressed?

Development of anti-DSG2 antibody-drug conjugates (ADCs) presents several technical challenges that researchers must navigate:

  • Antibody selection considerations:

    • Choose antibodies recognizing extracellular epitopes of DSG2 accessible on intact cells

    • Evaluate internalization efficiency, as effective ADCs require antibodies that trigger endocytosis

    • Select antibodies with high specificity to minimize off-target effects on tissues with low DSG2 expression

  • Conjugation chemistry optimization:

    • Determine optimal drug-to-antibody ratio (DAR) that maintains antibody binding while maximizing cytotoxic payload

    • Select appropriate linker chemistry (cleavable vs. non-cleavable) based on intracellular processing requirements

    • Validate that conjugation doesn't compromise antibody binding affinity or specificity

  • Payload selection strategy:

    • Match cytotoxic agent to target cell sensitivity (e.g., doxorubicin for proliferating cells)

    • Consider mechanism of action appropriate for the desired outcome (apoptosis induction for pluripotent stem cell elimination)

    • Evaluate payload stability in circulation vs. release kinetics in target cells

  • In vitro validation approach:

    • Establish assays comparing cytotoxicity in DSG2-high vs. DSG2-low/negative cells

    • Confirm mechanism of action using microscopy to track intracellular trafficking and drug release

    • Validate pH-dependent drug release in endosomal compartments

  • In vivo testing considerations:

    • Design animal models that recapitulate the target biology (e.g., stem cell xenografts)

    • Include appropriate controls (unconjugated antibody, non-targeting ADC)

    • Monitor biodistribution, pharmacokinetics, and potential off-target toxicities

  • Scale-up and reproducibility challenges:

    • Develop consistent conjugation protocols with well-defined quality control parameters

    • Establish stability testing under various storage conditions

    • Create analytical methods to assess batch-to-batch consistency

By systematically addressing these challenges, researchers can develop effective anti-DSG2 ADCs for applications such as eliminating pluripotent stem cells from therapeutic cell preparations, thus enhancing safety profiles of regenerative medicine approaches.

How do anti-DSG2 autoantibody levels in post-COVID-19 patients compare with those in ARVC and other cardiac conditions?

Comparative analysis of anti-DSG2 autoantibody levels across different cardiac conditions has revealed important insights:

  • Post-COVID-19 vs. healthy controls:

    • Post-COVID-19 patients show significantly elevated anti-DSG2 autoantibody levels compared to healthy individuals

    • Mean signal intensity: 19±83.2 in post-COVID-19 vs. 2.1±6.8 in healthy controls (P<0.001)

    • Approximately 29.3% of post-COVID-19 patients exhibit levels above the 90th percentile of the control population

  • Post-COVID-19 vs. ARVC patients:

    • About 8.7% of post-COVID-19 patients demonstrate anti-DSG2 autoantibody levels higher than the median found in diagnosed ARVC patients

    • This suggests a potential overlapping immunological mechanism between post-COVID cardiac manifestations and ARVC

  • Temporal stability in post-COVID-19 patients:

    • Longitudinal analysis shows no significant difference in signal intensity between samples collected at 6 months versus 9 months post-infection (p=0.529)

    • This persistence indicates a sustained autoimmune response rather than a transient phenomenon

  • ARVC specificity:

    • Anti-DSG2 autoantibodies appear to be specific for ARVC among primary cardiomyopathies

    • They are absent in healthy subjects and patients with other forms of cardiac disease, suggesting potential utility as a diagnostic biomarker

These comparative findings suggest that COVID-19 infection may trigger an autoimmune response targeting DSG2 that persists long after acute infection resolves. The similarity to ARVC-associated antibody profiles raises important questions about potential long-term cardiac effects of COVID-19 and suggests that monitoring anti-DSG2 autoantibodies might have prognostic value for identifying patients at risk for post-COVID cardiac complications.

What mechanisms might explain the generation of anti-DSG2 autoantibodies following viral infections?

The emergence of anti-DSG2 autoantibodies following viral infections, particularly COVID-19, likely involves several interrelated immunological mechanisms:

  • Epitope unmasking hypothesis:

    • Viral infection causes cardiac tissue damage, exposing previously hidden (cryptic) epitopes of DSG2

    • These normally sequestered epitopes become accessible to the immune system during tissue injury and repair

    • The exposed epitopes are recognized as foreign, triggering an autoimmune response

  • Molecular mimicry mechanisms:

    • Structural similarities between viral proteins and DSG2 epitopes may exist

    • T and B cells activated against viral antigens cross-react with self-antigens sharing sequence or structural homology

    • This cross-reactivity leads to autoantibody production targeting DSG2

  • Immune dysregulation pathways:

    • Viral infections, particularly COVID-19, can cause profound dysregulation of immune responses

    • Cytokine storms and aberrant T cell activation may compromise self-tolerance mechanisms

    • Breakdown of regulatory mechanisms leads to inappropriate activation of autoreactive B cells

  • Inflammation-mediated bystander activation:

    • Intense inflammatory environment during viral infection causes non-specific activation of immune cells

    • This "bystander activation" can trigger autoreactive B cells specific for DSG2

    • Persistent inflammation prolongs exposure to self-antigens, reinforcing autoimmune responses

  • Genetic susceptibility factors:

    • Individual genetic background influences susceptibility to autoimmunity

    • Polymorphisms in immune regulatory genes may predispose certain individuals to develop anti-DSG2 autoantibodies

    • This could explain why only a subset of infected individuals develop these autoantibodies

Understanding these mechanisms is essential for developing targeted interventions to prevent or mitigate autoimmunity-related cardiac complications following viral infections. Research into these pathways may also inform broader strategies for addressing virus-triggered autoimmune conditions affecting the heart.

How might the presence of anti-DSG2 antibodies influence the design of cardiac monitoring protocols for post-COVID-19 patients?

The discovery of persistent anti-DSG2 autoantibodies in post-COVID-19 patients has important implications for developing evidence-based cardiac monitoring protocols:

  • Risk stratification approach:

    • Screen recovered COVID-19 patients for anti-DSG2 autoantibodies, particularly focusing on those with high antibody levels (>90th percentile of control population)

    • Prioritize cardiac evaluation for the approximately 8.7% of patients with antibody levels comparable to those seen in ARVC patients

    • Consider more intensive monitoring for patients with sustained high levels at both 6 and 9 months post-infection

  • Multi-modal cardiac assessment:

    • Implement comprehensive cardiac evaluation including electrocardiography, echocardiography, and potentially cardiac MRI for patients with elevated anti-DSG2 autoantibodies

    • Monitor for ARVC-like manifestations including right ventricular dysfunction, ventricular arrhythmias, and conduction abnormalities

    • Correlate imaging findings with anti-DSG2 antibody levels to establish predictive associations

  • Extended monitoring timeline:

    • Given the documented persistence of anti-DSG2 autoantibodies at 9 months post-infection, extend cardiac monitoring beyond acute recovery phase

    • Implement longitudinal follow-up at 6, 12, and 24 months post-infection for patients with elevated antibody levels

    • Track potential progression or resolution of cardiac abnormalities relative to antibody dynamics

  • Integrated biomarker approach:

    • Combine anti-DSG2 autoantibody testing with established cardiac biomarkers (troponin, BNP) for comprehensive risk assessment

    • Develop integrated risk scores incorporating antibody levels, conventional biomarkers, and clinical parameters

    • Validate predictive models in prospective cohort studies

  • Intervention threshold determination:

    • Establish evidence-based thresholds for therapeutic intervention based on antibody levels and associated cardiac findings

    • Consider prophylactic treatments for high-risk patients, potentially including anti-inflammatory or immunomodulatory approaches

    • Design clinical trials to evaluate whether reducing anti-DSG2 autoantibody levels improves cardiac outcomes

Implementation of such protocols could enable early identification of patients at risk for long-term cardiac sequelae following COVID-19, potentially improving outcomes through timely intervention and targeted monitoring.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.