Biomarkers over Time: From Visual Contrast Sensitivity to Transcriptomics in Differentiating Chronic Inflammatory Response Syndrome and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome
Abstract
1. Introduction
2. Early Phase (1997–2004): The Neurotoxin Model and Visual Contrast Sensitivity
3. Formative Phase (2005–2010): Multisystem Biomarker Validation and Structured Case Definitions
4. Imaging and Neuroimmune Refinement (2010–2014)
5. Transcriptomic Era Begins (2015–2017): From RNA-Seq to Targeted Expression Profiling
6. Systems Integration (2017–2020): Diagnostic Convergence in Clinical Practice
7. Environmental Genomics and Causation (2021–22)
8. Transcriptomic Expansion and Neuroimmune Risk Signaling (2023–2024)
9. Ongoing Validation and Future Research (2025–)
10. Discussion
Funding
Acknowledgments
Conflicts of Interest
References
- Graves, B.S.; Patel, M.; Newgent, H.; Parvathy, G.; Nasri, A.; Moxam, J.; Gill, G.S.; Sawhney, V.; Gupta, M. Chronic Fatigue Syndrome: Diagnosis, Treatment, and Future Direction. Cureus 2024, 16, e70616. [Google Scholar] [CrossRef] [PubMed]
- Dooley, M.; Vukelic, A.; Jim, L. Chronic inflammatory response syndrome: A review of the evidence of clinical efficacy of treatment. Ann. Med. Surg. 2024, 86, 7248–7254. [Google Scholar] [CrossRef] [PubMed]
- Shoemaker, R.C.; Heyman, A.; Mancia, A.; Ryan, J.C. Inflammation Induced Chronic Fatiguing Illnesses: A steady march towards understanding mechanisms and identifying new biomarkers and therapies. Intern. Med. Rev. 2017, 3, 1–29. [Google Scholar]
- Grach, S.L.; Seltzer, J.; Chon, T.Y.; Ganesh, R. Diagnosis and Management of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Mayo Clin. Proc. 2023, 98, 1544–1551. [Google Scholar] [CrossRef]
- Okamoto, L.E.; Urechie, V.; Rigo, S.; Abner, J.J.; Giesecke, M.; Muldowney, J.A.S.; Furlan, R.; Shibao, C.A.; Shirey-Rice, J.K.; Pulley, J.M.; et al. Hyperadrenergic Postural Tachycardia Syndrome: Clinical Biomarkers and Response to Guanfacine. Hypertension 2024, 81, 2237–2247. [Google Scholar] [CrossRef]
- Clarke, K.S.P.; Kingdon, C.C.; Hughes, M.P.; Lacerda, E.M.; Lewis, R.; Kruchek, E.J.; Dorey, R.A.; Labeed, F.H. The search for a blood-based biomarker for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): From biochemistry to electrophysiology. J. Transl. Med. 2025, 23, 149. [Google Scholar] [CrossRef]
- Johnson, L.; Shapiro, M.; Needell, D.; Stricker, R.B. Optimizing Exclusion Criteria for Clinical Trials of Persistent Lyme Disease Using Real-World Data. Healthcare 2024, 13, 20. [Google Scholar] [CrossRef]
- Tsamou, M.; Kremers, F.A.C.; Samaritakis, K.A.; Roggen, E.L. Identifying microRNAs Possibly Implicated in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and Fibromyalgia: A Review. Int. J. Mol. Sci. 2024, 25, 9551. [Google Scholar] [CrossRef]
- Sun, Y.; Zhang, Z.; Qiao, Q.; Zou, Y.; Wang, L.; Wang, T.; Lou, B.; Li, G.; Xu, M.; Wang, Y.; et al. Immunometabolic changes and potential biomarkers in CFS peripheral immune cells revealed by single-cell RNA sequencing. J. Transl. Med. 2024, 22, 925. [Google Scholar] [CrossRef]
- Maya, J. Surveying the Metabolic and Dysfunctional Profiles of T Cells and NK Cells in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Int. J. Mol. Sci. 2023, 24, 11937. [Google Scholar] [CrossRef]
- Elahi, S.; Rezaeifar, M.; Osman, M.; Shahbaz, S. Exploring the role of galectin-9 and artemin as biomarkers in long COVID with chronic fatigue syndrome: Links to inflammation and cognitive function. Front. Immunol. 2024, 15, 1443363. [Google Scholar] [CrossRef]
- Azcue, N.; Tijero-Merino, B.; Acera, M.; Perez-Garay, R.; Fernandez-Valle, T.; Ayo-Mentxakatorre, N.; Ruiz-Lopez, M.; Lafuente, J.V.; Gomez Esteban, J.C.; Del Pino, R. Plasma Neurofilament Light Chain: A Potential Biomarker for Neurological Dysfunction in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Biomedicines 2024, 12, 1539. [Google Scholar] [CrossRef] [PubMed]
- Davis, L.; Higgs, M.; Snaith, A.; Lodge, T.A.; Strong, J.; Espejo-Oltra, J.A.; Kujawaki, S.; Zalewski, P.; Pretorius, E.; Hoerger, M.; et al. Dysregulation of lipid metabolism, energy production, and oxidative stress in myalgic encephalomyelitis/chronic fatigue syndrome, Gulf War Syndrome and fibromyalgia. Front. Neurosci. 2025, 19, 1498981. [Google Scholar] [CrossRef]
- Huang, K.; Lidbury, B.A.; Thomas, N.; Gooley, P.R.; Armstrong, C.W. Machine learning and multi-omics in precision medicine for ME/CFS. J. Transl. Med. 2025, 23, 68. [Google Scholar] [CrossRef] [PubMed]
- Xiong, R.; Aiken, E.; Caldwell, R.; Vernon, S.D.; Kozhaya, L.; Gunter, C.; Bateman, L.; Unutmaz, D.; Oh, J. BioMapAI: Artificial Intelligence Multi-Omics Modeling of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. bioRxiv 2025. [Google Scholar] [CrossRef]
- Yagin, F.; Alkhateeb, A.; Raza, A.; Samee, N.; Mahmoud, N.; Colak, C.; Yagin, B. An Explainable Artificial Intelligence Model Proposed for the Prediction of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and the Identification of Distinctive Metabolites. Diagnostics 2023, 13, 3495. [Google Scholar] [CrossRef]
- Thomas, N.; Chau, T.; Tantanis, D.; Huang, K.; Scheinberg, A.; Gooley, P.R.; Josev, E.K.; Knight, S.J.; Armstrong, C.W. Serial Paediatrics Omics Tracking in Myalgic Encephalomyelitis (SPOT-ME): Protocol paper for a multidisciplinary, observational study of clinical and biological markers of paediatric myalgic encephalomyelitis/chronic fatigue syndrome in Australian adolesc. BMJ Open 2024, 14, e089038. [Google Scholar] [CrossRef]
- Smyth, N.J.; Blitshteyn, S. Language Matters: What Not to Say to Patients with Long COVID, Myalgic Encephalomyelitis/Chronic Fatigue Syndrome, and Other Complex Chronic Disorders. Int. J. Environ. Res. Public Health 2025, 22, 275. [Google Scholar] [CrossRef]
- Nunes, M.; Vlok, M.; Proal, A.; Kell, D.B.; Pretorius, E. Data-independent LC-MS/MS analysis of ME/CFS plasma reveals a dysregulated coagulation system, endothelial dysfunction, downregulation of complement machinery. Cardiovasc. Diabetol. 2024, 23, 254. [Google Scholar] [CrossRef]
- Huang, K.; GC de Sá, A.; Thomas, N.; Phair, R.D.; Gooley, P.R.; Ascher, D.B.; Armstrong, C.W. Discriminating Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and comorbid conditions using metabolomics in UK Biobank. Commun. Med. 2024, 4, 248. [Google Scholar] [CrossRef]
- Shoemaker, R.C.; Johnson, K.; Jim, L.; Berry, Y.; Dooley, M.; Ryan, J.C.; McMahon, S.W. Diagnostic Process for Chronic Inflammatory Response Syndrome (CIRS): A Consensus Statement Report of the Consensus Committee of Surviving Mold. Intern. Med. Rev. 2018, 4, 1–47. [Google Scholar]
- McMahon, S.W. An Evaluation of Alternate Means to Diagnose Chronic Inflammatory Response Syndrome and Determine Prevalence. Med. Res. Arch. 2017, 5, 1–17. [Google Scholar] [CrossRef]
- Shoemaker, R.C.; Hudnell, H.K. Possible Estuary-Associated Syndrome: Symptoms, Vision, and Treatment. Environ. Health Perspect. 2001, 109, 539–545. [Google Scholar] [CrossRef] [PubMed]
- Shoemaker, R.C.; Rash, J.M.; Simon, E.W. Sick Building Syndrome in water-damaged buildings: Generalization of the chronic biotoxin-associated illness paradigm to indoor toxigenic fungi. In Bioaerosols, Fungi, Bacteria, Mycotoxins and Human Health; Johanning, E., Ed.; The Fungal Research Group Foundation: Albany, NY, USA, 2005; pp. 66–67. [Google Scholar]
- Shoemaker, R.C.; House, D.E. Sick building syndrome (SBS) and exposure to water-damaged buildings: Time series study, clinical trial and mechanisms. Neurotoxicol. Teratol. 2006, 28, 573–588. [Google Scholar] [CrossRef] [PubMed]
- Shoemaker, R.C. Suriving Mold Life in the Era of Dangerous Buildings; Otter Bay Books, LLC: Baltimore, MD, USA, 2010. [Google Scholar]
- Shoemaker, R.C.; House, D.; Ryan, J.C. Vasoactive intestinal polypeptide (VIP) corrects chronic inflammatory response syndrome (CIRS) acquired following exposure to water-damaged buildings. Health 2013, 5, 396–401. [Google Scholar] [CrossRef]
- Shoemaker, R.C.; House, D.; Ryan, J.C. Structural brain abnormalities in patients with inflammatory illness acquired following exposure to water-damaged buildings: A volumetric MRI study using NeuroQuant(R). Neurotoxicol. Teratol. 2014, 45, 18–26. [Google Scholar] [CrossRef]
- Ryan, J.C.; Wu, Q.; Shoemaker, R.C. Transcriptomic signatures in whole blood of patients who acquire a chronic inflammatory response syndrome (CIRS) following an exposure to the marine toxin ciguatoxin. BMC Med. Genom. 2015, 8, 15. [Google Scholar] [CrossRef]
- Ryan, J.C.; Shoemaker, R.C. RNA-Seq on patients with chronic inflammatory response syndrome (CIRS) treated with vasoactive intestinal peptide (VIP) shows a shift in metabolic state and innate immune functions that coincide with healing. Med. Res. Arch. 2016, 4, 1–10. [Google Scholar]
- McMahon, S.W.; Shoemaker, R.C.; Ryan, J.C. Reduction in Forebrain Parenchymal and Cortical Grey Matter Swelling Across Treatment Groups in patients with inflammatory illness. J. Neurosci. Clin. Res. 2016, 1, 2. Available online: https://www.scitechnol.com/peer-review/reduction-in-forebrain-parenchymal-and-cortical-grey-matter-swelling-across-treatment-groups-in-patients-with-inflammatory-illness-RoPB.php?article_id=4804 (accessed on 11 July 2025). [CrossRef]
- Shoemaker, R.C.; McMahon, S.W.; Heyman, A.; Lark, D.; M., v.d.W.; Ryan, J.C. Treatable metabolic and inflammatory abnormalities in Post COVID Syndrome (PCS) define the transcriptomic basis for persistent symptoms: Lessons from CIRS. Med. Res. Arch. 2021, 9, 1–18. [Google Scholar] [CrossRef]
- Shoemaker, R.C.; Katz, D.; Ackerly, M.; Rapaport, S.; McMahon, S.W.; Berndtson, K.; Ryan, J.C. Intranasal VIP safely restores volume to multiple grey matter nuclei in patients with CIRS. Intern. Med. Rev. 2017, 3, 1–14. [Google Scholar] [CrossRef]
- Shoemaker, R.C. Metabolism, molecular hypometabolism and inflammation: Complications of proliferative physiology include metabolic acidosis, pulmonary hypertension, T reg cell deficiency, insulin resistance and neuronal injury. Trends Diabetes Metab. 2020, 3, 1–15. [Google Scholar] [CrossRef]
- Shoemaker, R.C.; Neil, V.; Heyman, A.; van der Westhuizen, M.; McMahon, S.W.; Lark, D. New Molecular Methods Bring New Insight into Human- And Building -Health Risk Assessments from Water-Damaged Buildings: Defining Exposure and Reacity, the Two Sides of Causation of CIRS-WDB Illness. Med. Res. Arch. 2021, 9, 1–36. [Google Scholar] [CrossRef]
- Shoemaker, R.; Heyman, A.; Lark, D. TUBB1, TUBA4A and MAPK as Indicators of Die-Back Degenerative Central Nervous System Disease in Patients Sickened by Specific Exposure to the Interior Environment of Water- Damaged Buildings. Med. Res. Arch. 2023, 11, 1–9. [Google Scholar] [CrossRef]
- Shoemaker, R.C.; Heyman, A.; Lark, D. Exposure to the Interior Environment of Water-Damaged Buildings Can Activate HIF 1A, Induce Proliferative Physiology and Impair Mitochondrial Metabolism. Med. Res. Arch. 2024, 12, 1–11. [Google Scholar] [CrossRef]
- Shoemaker, R.C.; Ryan, J.C.; Heyman, A.; Dorninger, E.; McMahon, S.W.; Lark, D. A Transcriptomic Fingerprint for Parkinsons Disease Found in Patients with Chronic Inflammatory Response Syndrome: Implications for Diagnosis, Treatment and Prevention. Med. Res. Arch. 2024, 12, 1–9. [Google Scholar] [CrossRef]
- Shoemaker, R.C. Diagnosis of Pfiesteria-human illness syndrome. Md. Med. J. 1997, 1997, 521–523. [Google Scholar]
- Grattan, L.M.; Oldach, D.; Perl, T.M.; Lowitt, M.H.; Matuszak, D.L.; Dickson, C.; Parrott, C.; Shoemaker, R.C.; Kauffman, C.L.; Wasserman, M.P.; et al. Learning and memory difficulties after environmental exposure to waterways containing toxin-producing Pfiesteria or Pfiesteria-like dinoflagellates. Lancet 1998, 352, 532–539. [Google Scholar] [CrossRef]
- Shoemaker, R.C. Residential and Recreational Acquisition of Possible Estuary-Associated Syndrome: A New Approach to Successful Diagnosis and Treatment. Environ. Health Perspect. 2001, 109, 791–796. [Google Scholar]
- United States Government Accountability Office (GAO). Indoor Mold: Better Coordination of Research on Health Effects and More Consistent Guidance Would Improve Federal Efforts. Report to the Chairman, Committee on Health, Education, Labor and Pensions, U.S. Senate. GAO-08-980. U.S. Government Accountability Office: Washington, DC, USA, September 2008. Available online: https://www.gao.gov/assets/gao-08-980.pdf (accessed on 29 November 2019).
- Hosainzadegan, H.; Khalilov, R.; Gholizadeh, P. The necessity to revise Koch’s postulates and its application to infectious and non-infectious diseases: A mini-review. Eur. J. Clin. Microbiol. Infect. Dis. 2020, 39, 215–218. [Google Scholar] [CrossRef]
- Shoemaker, R.C.; House, D.E. A time-series study of sick building syndrome: Chronic, biotoxin-associated illness from exposure to water-damaged buildings. Neurotoxicol. Teratol. 2005, 27, 29–46. [Google Scholar] [CrossRef]
- ProgeneDx. FAQS for GENIE testing. Available online: https://www.progenedx.com/faqs (accessed on 11 July 2025).
- Shoemaker, R.C.; Heyman, A.; Lark, D. Transcriptomics and Brain Volumetrics Define the Causes of Cognitive Impairment in Patients with CIRS and Support the use of VIP in Treatment. Med. Res. Arch. 2023, 11, 1–7. [Google Scholar] [CrossRef]
- Bouquet, J.; Soloski, M.J.; Swei, A.; Cheadle, C.; Federman, S.; Billaud, J.-N.; Rebman, A.W.; Kabre, B.; Halpert, R.; Boorgula, M.; et al. Longitudinal Transcriptome Analysis Reveals a Sustained Differential Gene Expression Signature in Patients Treated for Acute Lyme Disease. mBio 2016, 7, e00100-16. [Google Scholar] [CrossRef]
- Shoemaker, R.C.; Lark, D. Screening for Biomarkers of Actinobacteria Associated with Water—Damaged Buildings—Part 1. Med. Res. Arch. 2021, 10, 1–21. [Google Scholar] [CrossRef]
- Shoemaker, R.C.; Meinhardt, J.; Heyman, A.; Lark, D. Exposure to Actinobacteria resident in water-damaged buildings and resultant immune injury in Chronic Inflammatory Response Syndrome. Med. Res. Arch. 2021, 9, 1–28. [Google Scholar] [CrossRef]
Feature | CIRS | ME/CFS |
---|---|---|
Pathogenesis | Innate immune dysregulation triggered primarily by environmental or biotoxin exposure | Unclear; proposed mitochondrial, immune, infectious, and neuroinflammatory roles |
Diagnosis | Structured case definition using symptoms, biomarkers, and treatment response | Symptom-based case definitions; diagnosis of exclusion |
Validated Biomarkers | Yes—defined and reproducible biomarker panel | No clinically validated biomarker panel |
Reproducibility | High—reproducible patterns in diagnostic biomarkers and consistent treatment-response normalization across cohorts | Low—omics findings often not reproducible across cohorts |
Treatment Response | Protocol-based; objective improvements in labs and symptoms documented | No curative treatment; management is supportive |
Case Definition Origin | Biomarker-guided and treatment-responsive; refined over two decades | Multiple symptom-based definitions; heterogeneity persists |
Symptom Overlap | Fatigue, cognitive dysfunction, post-exertional symptoms, autonomic dysregulation | Similar symptoms; post-exertional malaise (PEM) emphasized in recent definitions |
Biomarker/Concept | Category | First Publication | Diagnostic Relevance | Type of Study |
---|---|---|---|---|
Visual contrast sensitivity (VCS) | Functional/Screening | Environmental Health Perspectives, 2001 [23] | Earliest objective test used in neurotoxic illness; confirmed reproducible in Possible Estuary-Associated Syndrome (PEAS) | Case series (5 patients) |
Human leukocyte antigen DR/DQ (HLA DR/DQ) haplotypes | Genetic susceptibility | Bioaerosols, 2005 [24] | Used to determine risk for biotoxin illness across mold, Lyme, and dinoflagellate exposures | Case–control (156 cases/111 controls) |
Melanocyte stimulating hormone (MSH) | Neuroendocrine | Bioaerosols, 2005 [24] | Key regulatory peptide; consistently low in CIRS patients | Case–control (156 cases/111 controls) |
Matrix metalloproteinase 9 (MMP-9) | Inflammatory | Bioaerosols, 2005 [24] | Elevated in inflammatory response; used to monitor response to treatment | Case–control (156 cases/111 controls) |
Multiple antibiotic-resistant coagulase-negative staphylococci (MARCoNS) | Infectious/Inflammatory | Bioaerosols, 2005 [24] | Associated with low MSH, persistent inflammation, and biofilm formation; eradication improves clinical and biomarker outcomes | Case–control (156 cases/111 controls) |
Leptin | Metabolic | Bioaerosols, 2005 [24] | Elevated in CIRS; drops with therapy | Case–control (156 cases/111 controls) |
Adrenocorticotropic hormone (ACTH)/Cortisol | Neuroendocrine | Bioaerosols, 2005 [24] | HPA axis dysregulation | Case–control (156 cases/111 controls) |
antidiuretic hormone (ADH)/Osmolality | Hormonal/Fluid Balance | Bioaerosols, 2005 [24] | Volume dysregulation in MSH-deficient states | Case–control (156 cases/111 controls) |
First formal case definition for Chronic Biotoxin Associated Illness (CBAI) | Diagnostic Framework | Bioaerosols, 2005 [24] | Exposure + symptoms + biomarkers + response | Case–control (156 cases/111 controls) |
Vascular endothelial growth factor (VEGF) | Perfusion/Hypoxia | Neurotoxicology and Teratology, 2006 [25] | Biphasic; abnormal regulation indicates hypoxia, low capillary perfusion | Case time series (28 patients); randomized controlled trial (13 patients) |
Complement component 4a C4a | Complement activation | Surviving Mold, 2010 [26] | Innate immune activation marker; rises with re-exposure | Clinical observation (uncontrolled; data compiled in Surviving Mold) |
Vasoactive Intestinal Peptide (VIP) | Neuroendocrine | Surviving Mold, 2010 [26] | Key regulatory peptide | Clinical observation (uncontrolled; data compiled in Surviving Mold) |
TGF-β1 | Fibrosis/Cytokine | Surviving Mold, 2010 [26] | Pro-fibrotic cytokine; key inflammatory marker in CIRS | Clinical observation (uncontrolled; data compiled in Surviving Mold) |
VIP | Neuropeptide/Therapeutic | Health, 2013 [27] | Restores immune regulation; corrects many CIRS abnormalities | Case–control (156 cases/111 controls) |
NeuroQuant® (Cortech Labs, Inc., San Diego, CA, USA) MRI | Neuroimaging | Neurotoxicology and Teratology, 2014 [28] | Quantifies grey matter changes (e.g., caudate atrophy); reversible with treatment | Case–control (17 cases/18 controls) |
Caudate, hippocampus, thalamus, pallidum, putamen, cerebellum | Neuroimaging/Volumetric | Neurotoxicology and Teratology, 2014 [28] | NeuroQuant volumetric targets used to track structural brain changes associated with CIRS; reversible with VIP treatment | Case–control (17 case/18 controls) |
Transcriptomic fingerprint (microarray) | Transcriptomics | BMC Medical Genomics, 2015 [29] | First transcriptomic classification of CIRS triggered by ciguatera toxin; clear gene pattern | Case–control (11 cases/11 controls) |
RNA-Sequencing (RNA-Seq) post-VIP | Transcriptomics/Response | Medical Research Archives, 2016 [30] | Showed downregulation of ribosomal and mitochondrial genes after VIP | Prospective observational (14 cases) |
IKZF1 and VIPR1 | Transcriptomic/Regulatory | Medical Research Archives, 2016 [30] | Downregulation linked to poor response to VIP and increased grey matter atrophy | Prospective observational (14 cases) |
NeuroQuant reversal with treatment | Neuroimaging/Response | Journal of Neuroscience & Clinical Research, 2016 [31] | Grey matter and forebrain swelling reversed with treatment | Case–control (28 cases/23 controls) |
Symptom clusters (13) | Clinical Screening | Internal Medicine Review, 2017 [32] | Diagnostic tool; ≥8/13 clusters predictive of CIRS | Retrospective observational analysis of clinical symptom data (n > 1000) |
VIP-integrated imaging/lab study | Therapeutic Systems Integration | Internal Medicine Review, 2017 [33] | Showed lab normalization, grey matter volume restoration, and gene shift | Open-label trial (35 patients) |
Translocase | Transcriptomic/Metabolic | Trends in Diabetes and Metabolism, 2020 [34] | Downregulated in proliferative physiology; contributes to altered pyruvate handling and reduced mitochondrial adenosine triphosphate (ATP) production | Cross-sectional cohort analysis (n = 112 consecutive Gene Expression: Inflammation Explained (GENIE) Stage 1 patients) |
IRS2 | Transcriptomic/Metabolic | Trends in Diabetes and Metabolism, 2020 [34] | Upregulated in proliferative physiology; indicates intracellular insulin resistance and altered glucose metabolism | Cross-sectional cohort analysis (n = 112 consecutive GENIE Stage 1 patients) |
Molecular hypometabolism (MHM) | Transcriptomics/Diagnostic Classifier | Trends in Diabetes and Metabolism, 2020 [34] | Ribosomal and mitochondrial gene suppression; a core transcriptomic pattern in CIRS | Cross-sectional cohort analysis (n = 112 consecutive GENIE Stage 1 patients) |
GENIE causation model | Causation/Transcriptomic Staging | Medical Research Archives, 2021 [35] | Defines stage-based diagnostic thresholds using GENIE + environmental exposure | Retrospective observational study with stage-stratified transcriptomic and environmental analysis (n ≈ 238 across Stages 0–5) |
Actinobacteria | Environmental Trigger (NGS) | Medical Research Archives, 2021 [35] | First paper to link Actinobacterial presence in WDB environments with human transcriptomic signatures of inflammation (e.g., MAPK1, TGFBR1); supports gene-environment causation framework in CIRS | Retrospective observational study with stage-stratified transcriptomic and environmental analysis (n ≈ 238 across Stages 0–5) |
Post-Covid Syndrome (PCS) vs. CIRS | Transcriptomic Comparison | Medical Research Archives, 2021 [32] | PCS patients with CIRS features showed MHM, CD3D suppression, TGFBR upregulation | Prospective observational study (n = 21 PCS patients; proof-of-concept transcriptomic analysis) |
Actino Skin® and Actino Plasma® | Translational/Diagnostic | Commercial product; patent pending ≈ 2022 | Skin—quantitative polymerase chain reaction (qPCR)-based test for Human Habitat (HH)Actinobacteria on skin; supports exposure assessment in CIRS Plasma—quantifies immune reactivity to Actinobacterial mycolic acids; reflects systemic response in CIRS | Commercial assay development (patent pending; preliminary internal validation) |
TUBB1, TUBA4A, Mitogen-associated protein kinase (MAPK) | Transcriptomic/Neurodegeneration | Medical Research Archives, 2023 [36] | Proposed markers for CNS injury (caudate atrophy) in CIRS | Retrospective observational study (386 of 1822 GENIE tests) |
HIF 1A | Transcriptomic/Metabolic | Medical Research Archives, 2024 [37] | Upregulated following WDB exposure; reflects proliferative physiology marked by impaired mitochondrial metabolism, increased glycolysis, and heightened inflammatory signaling in CIRS | Retrospective observational study (81 of 1822 GENIE tests) |
CLU, GP6, GP9, PF4, ITGA2B | Transcriptomic/Neuroimmune-Coagulation | Medical Research Archives, 2024 [38] | Co-expression of these genes defines the “triple-positive neuroimmune risk profile” in CIRS, associated with caudate atrophy, cytoskeletal disruption, and poor VIP response; overlaps with Parkinson’s disease transcriptomic signatures | Case series (77 patients); validation cohort (102 consecutive CIRS patients: 36 Triple Positives, 66 controls); retrospective analysis (171 of 1822 GENIE tests) |
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. |
© 2025 by the author. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Dooley, M. Biomarkers over Time: From Visual Contrast Sensitivity to Transcriptomics in Differentiating Chronic Inflammatory Response Syndrome and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Int. J. Mol. Sci. 2025, 26, 7284. https://doi.org/10.3390/ijms26157284
Dooley M. Biomarkers over Time: From Visual Contrast Sensitivity to Transcriptomics in Differentiating Chronic Inflammatory Response Syndrome and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. International Journal of Molecular Sciences. 2025; 26(15):7284. https://doi.org/10.3390/ijms26157284
Chicago/Turabian StyleDooley, Ming. 2025. "Biomarkers over Time: From Visual Contrast Sensitivity to Transcriptomics in Differentiating Chronic Inflammatory Response Syndrome and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome" International Journal of Molecular Sciences 26, no. 15: 7284. https://doi.org/10.3390/ijms26157284
APA StyleDooley, M. (2025). Biomarkers over Time: From Visual Contrast Sensitivity to Transcriptomics in Differentiating Chronic Inflammatory Response Syndrome and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. International Journal of Molecular Sciences, 26(15), 7284. https://doi.org/10.3390/ijms26157284