Genetic inflammatory signature defines depression subtypes and treatment response

Researchers led by Prof. Alessandro Serretti at Kore University of Enna have identified a genetic inflammatory signature that defines specific depression subtypes and influences how patients respond to antidepressant medications, according to new peer-reviewed research published today in Genomic Psychiatry. The findings suggest that inherited predisposition to inflammation may help explain why certain patients experience particular symptom patterns and respond differently to standard treatments, potentially advancing efforts toward more personalized approaches in mental health care.

Novel genetic architecture uncovered

The research team analyzed polygenic scores for C-reactive protein, a key marker of inflammation, in 1059 European patients with major depressive disorder who received at least four weeks of antidepressant treatment. Using advanced genetic scoring methods derived from UK Biobank data encompassing over 223,000 individuals, the investigators discovered that genetic liability for elevated CRP correlates with distinct clinical features including increased body mass index, altered appetite regulation, and specific patterns of treatment response.

The polygenic scores were computed using L1-penalized regression weights through the snpnet algorithm, incorporating approximately 1.08 million genetic variants. This sophisticated approach achieved a robust predictive capacity with an R² of 0.1215 for log-transformed CRP levels in independent test samples. The methodology represents a significant advance in capturing complex genetic architecture underlying inflammatory processes in psychiatric conditions.

Prof. Serretti and colleagues found that patients with higher CRP polygenic scores demonstrated less weight and appetite loss following treatment (r = -0.07, p = 0.02 for weight loss; r = -0.06, p = 0.044 for appetite reduction), earlier age of depression onset (mean difference of approximately 2 years, p = 0.046), and lower employment status (r = -0.06, p = 0.047). These associations remained significant even after accounting for overall depression severity, suggesting that inflammatory genetic predisposition shapes a specific constellation of symptoms rather than simply increasing overall illness burden.

Unexpected treatment response patterns challenge conventional understanding

The study revealed a surprising U-shaped relationship between CRP genetic liability and antidepressant outcomes. Treatment-resistant patients showed the highest polygenic scores, followed unexpectedly by treatment responders, while nonresponders demonstrated the lowest scores (F = 3.52, p = 0.03). This nonlinear pattern persisted even after controlling for established clinical predictors including age, episode frequency, suicidal ideation, anxiety comorbidity, employment status, functional disability scores, antipsychotic augmentation, illness duration, and previous treatment trials.

Statistical analysis using generalized linear models confirmed the quadratic relationship, with the quadratic term achieving statistical significance (β = 0.16, p = 0.013). When stratified into quintiles, the probability of nonresponse was highest in the lowest CRP-PGS quintile and declined thereafter, while both responder and treatment-resistant depression probabilities showed progressive increases at higher quintiles. Bootstrap 95% confidence intervals validated the robustness of these unexpected patterns.

When incorporated into multivariable models, the CRP polygenic score demonstrated even stronger association with treatment outcomes (F = 7.69, p < 0.001), explaining an additional 1.9% of variance beyond conventional clinical predictors. While this effect size appears modest, it represents independent prognostic information not captured by traditional staging approaches, potentially identifying patients who might benefit from alternative or augmented treatment strategies.

Depression as a global health challenge requiring new approaches

Major depressive disorder affects over 280 million people worldwide and represents one of the leading causes of disability globally. Despite decades of research and numerous available treatments, approximately 30% of patients fail to achieve adequate remission with standard therapies, and up to 15% develop treatment-resistant depression. This heterogeneity in treatment response has long frustrated clinicians and researchers alike, suggesting that depression may comprise multiple biological subtypes requiring different therapeutic approaches.

The concept of immunometabolic depression has emerged from converging lines of evidence showing that approximately one-quarter of depressed patients exhibit elevated inflammatory markers. These patients often present with distinct clinical features including increased somatic symptoms, cognitive dysfunction, metabolic disturbances, and differential treatment responses. The current findings provide genetic validation for this clinical observation, demonstrating that inherited variation in inflammatory pathways contributes to these phenotypic differences.

From historical observations to molecular understanding

The findings gain additional context through an accompanying editorial in Genomic Psychiatry that explores how these molecular discoveries validate clinical observations dating back to the 1897 French monograph "La Mélancolie" by Roubinovitch and Toulouse. The editorial authors, Dr. Julio Licinio and Dr. Ma-Li Wong, note that what 19th-century physicians described as "psychophysical decrease" and "distressing affective tone" may reflect the same immunometabolic processes now being elucidated through genetic research.

The editorial highlights how Roubinovitch and Toulouse documented detailed phenomenology including changes in "coenesthesia" (body feeling) that produced distressing affective tones, observations that remarkably parallel modern findings linking inflammatory genetics to somatic symptoms. Their meticulous documentation of 22 case histories, now available for the first time in English translation as supplementary material, reveals clinical insights that anticipated current understanding of depression heterogeneity by over a century.

This historical perspective underscores how careful phenomenological observation can capture biological truths that await molecular discovery. The consistency of depressive symptoms across centuries, now partly explained through inflammatory genetic architecture, suggests that combining traditional clinical wisdom with modern genomic approaches may yield deeper understanding of psychiatric conditions.

Mechanisms linking inflammation to depression

The biological pathways connecting CRP genetics to depression likely involve multiple interconnected systems. The genetic variants influencing CRP levels are enriched in hepatic endoplasmic reticulum stress pathways, IL-6/JAK-STAT signaling cascades, and lipid metabolism networks. These same pathways regulate neurotransmitter synthesis, hypothalamic-pituitary-adrenal axis function, and neural plasticity mechanisms critical for mood regulation.

Recent evidence suggests that peripheral inflammation can disrupt brain function through multiple routes including altered tryptophan metabolism, increased blood-brain barrier permeability, microglial activation, and disrupted reward processing circuits. The nonlinear relationship observed between CRP polygenic scores and treatment response may reflect complex interactions between these pathways, where moderate inflammation impairs serotonergic function while very high or very low levels engage compensatory mechanisms or alternative neurotransmitter systems.

Implications for precision psychiatry and treatment selection

The identification of an immunometabolic depression subtype has immediate implications for treatment development and patient stratification. Previous trials have shown that patients with elevated inflammatory markers may preferentially respond to anti-inflammatory augmentation strategies. The infliximab proof-of-concept study demonstrated that patients with baseline high-sensitivity CRP above 5 mg/L achieved approximately 4-point greater improvement on depression rating scales compared to placebo. Similar findings have emerged for other immunomodulatory interventions including minocycline, celecoxib, and omega-3 fatty acids.

The current findings suggest that germline genetic testing could help identify individuals likely to maintain elevated inflammation even during clinical remission, potentially guiding prophylactic or maintenance treatment decisions. Patients with high CRP polygenic scores might benefit from early augmentation with anti-inflammatory agents, lifestyle interventions targeting metabolic health, or alternative antidepressants with immunomodulatory properties.

Prof. Serretti emphasized that while polygenic scores remain population-level probabilistic tools rather than deterministic individual tests, they may contribute to multi-level assessment strategies incorporating both genetic predisposition and current inflammatory status. Integration with other biomarkers including circulating cytokines, neuroimaging markers of neuroinflammation, and metabolomic profiles could improve predictive accuracy sufficiently for clinical implementation.

Research methodology and multicenter validation

The study utilized data from the European Group for the Study of Resistant Depression (GSRD), a multicenter consortium collecting standardized clinical and genetic information across Austria, Belgium, France, Germany, Greece, Israel, Italy, and Switzerland. Participants underwent comprehensive assessment including the Montgomery-Åsberg Depression Rating Scale, Hamilton Depression Rating Scale, Sheehan Disability Scale, Mini-International Neuropsychiatric Interview, and detailed treatment history documentation.

All patients were naturalistically treated with at least one antidepressant at adequate doses for minimum four weeks, with treatment response defined as 50% or greater reduction in MADRS scores from baseline. Treatment resistance was classified according to established criteria as failure to respond to two or more adequate antidepressant trials. The naturalistic design, while introducing treatment heterogeneity, enhances real-world applicability of findings.

Genetic analysis employed state-of-the-art imputation methods using Haplotype Reference Consortium data and penalized regression techniques to maximize predictive accuracy while controlling for population stratification through principal component analysis. Quality control procedures removed variants with minor allele frequency below 0.01, poor imputation quality scores below 0.30, and genotype probability below 0.90, ensuring robust genetic associations.

Future research directions and clinical translation

The research team outlined several priority areas for future investigation. Prospective studies tracking inflammatory markers and clinical outcomes longitudinally could clarify causal relationships between genetic liability, current inflammation, and treatment trajectories. Gene-by-environment interaction studies examining how social adversity, childhood trauma, or medical comorbidities modify the expression of inflammatory genetic risk could identify modifiable factors for intervention.

Integration of CRP polygenic scores with other biological markers represents another promising avenue. Combining genetic data with peripheral biomarkers, neuroimaging signatures, and digital phenotyping could create comprehensive risk algorithms approaching clinically actionable accuracy. Machine learning approaches may identify complex patterns invisible to traditional statistical methods, potentially revealing subtypes within the immunometabolic depression spectrum.

The development of inflammation-guided treatment protocols requires careful consideration of both efficacy and safety. While anti-inflammatory augmentation shows promise for selected patients, indiscriminate use could potentially interfere with beneficial inflammatory processes including neuroplasticity and stress adaptation. Precision medicine approaches must balance targeting pathological inflammation while preserving physiological immune function.

Limitations and need for diverse population studies

The research team acknowledged important limitations requiring consideration. The cross-sectional design prevents definitive causal inference about whether inflammatory genetics drives symptom development or influences treatment response through independent mechanisms. The naturalistic treatment setting, while enhancing generalizability, introduces heterogeneity in drug selection, dosing, and adherence that may obscure specific pharmacogenetic interactions.

The exclusively European ancestry sample represents a critical limitation given known population differences in both genetic architecture and inflammatory processes. Replication in African, Asian, and admixed populations is essential before clinical implementation. Additionally, the study did not measure peripheral inflammatory markers, preventing direct comparison between genetic predisposition and current inflammatory status.

Statistical power for detecting gene-by-treatment interactions remained limited despite the relatively large sample size. Multiple testing across numerous clinical variables raises possibility of false positive findings, though the consistency with prior research supports validity of main results. Effect sizes, while statistically significant, remain modest from a clinical prediction standpoint, emphasizing need for integration with additional biomarkers.

This peer-reviewed research represents a significant advance in psychiatric genetics, offering new insights into depression heterogeneity through rigorous experimental investigation. The findings challenge existing paradigms regarding uniform treatment approaches for understanding major depressive disorder. By employing innovative polygenic scoring methodology, the research team has generated data that not only advances fundamental knowledge but also suggests practical applications in clinical psychiatry. The reproducibility and validation of these findings through the peer-review process ensures their reliability and positions them as a foundation for future investigations. This work exemplifies how cutting-edge research can bridge the gap between basic science and translational applications, potentially impacting treatment selection strategies in the coming years.

Source:
Journal reference:

Serretti, A., et al. (2025) Polygenic liability to C-reactive protein defines immunometabolic depression phenotypes and influences antidepressant therapeutic outcomes. Genomic Psychiatry. doi.org/10.61373/gp025r.0092

Comments

The opinions expressed here are the views of the writer and do not necessarily reflect the views and opinions of News Medical.
Post a new comment
Post

While we only use edited and approved content for Azthena answers, it may on occasions provide incorrect responses. Please confirm any data provided with the related suppliers or authors. We do not provide medical advice, if you search for medical information you must always consult a medical professional before acting on any information provided.

Your questions, but not your email details will be shared with OpenAI and retained for 30 days in accordance with their privacy principles.

Please do not ask questions that use sensitive or confidential information.

Read the full Terms & Conditions.

You might also like...
Low-calorie diets linked to higher psoriatic arthritis risk, genetic study suggests