1
|
Germain A, Jaycox JR, Emig CJ, Ring AM, Hanson MR. An In-Depth Exploration of the Autoantibody Immune Profile in ME/CFS Using Novel Antigen Profiling Techniques. Int J Mol Sci 2025; 26:2799. [PMID: 40141440 PMCID: PMC11943395 DOI: 10.3390/ijms26062799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating disorder characterized by serious physical and cognitive impairments. Recent research underscores the role of immune dysfunction, including the role of autoantibodies, in ME/CFS pathophysiology. Expanding on previous studies, we analyzed 7542 antibody-antigen interactions in ME/CFS patients using two advanced platforms: a 1134 autoantibody Luminex panel from Oncimmune and Augmenta Bioworks, along with Rapid Extracellular Antigen Profiling (REAP), a validated high-throughput method that measures autoantibody reactivity against 6183 extracellular human proteins and 225 human viral pathogen proteins. Unlike earlier reports, our analysis of 172 participants revealed no significant differences in autoantibody reactivities between ME/CFS patients and controls, including against GPCRs such as β-adrenergic receptors. However, subtle trends in autoantibody ratios between male and female ME/CFS subgroups, along with patterns of herpesvirus reactivation, suggest the need for broader and more detailed exploration.
Collapse
Affiliation(s)
- Arnaud Germain
- Department of Molecular Biology and Genetics, Biotechnology Building, Cornell University, Ithaca, NY 14853, USA;
| | - Jillian R. Jaycox
- Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (J.R.J.); (A.M.R.)
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Aaron M. Ring
- Division of Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (J.R.J.); (A.M.R.)
| | - Maureen R. Hanson
- Department of Molecular Biology and Genetics, Biotechnology Building, Cornell University, Ithaca, NY 14853, USA;
| |
Collapse
|
2
|
Lv C, Chen J, Wang Y, Lin Y. Immunoregulatory role of exosomal circRNAs in the tumor microenvironment. Front Oncol 2025; 15:1453786. [PMID: 40034598 PMCID: PMC11872884 DOI: 10.3389/fonc.2025.1453786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 01/02/2025] [Indexed: 03/05/2025] Open
Abstract
As cancer incidence and mortality rates rise, there is an urgent need to develop effective immunotherapy strategies. Circular RNA (circRNA), a newly identified type of non-coding RNA, is abundant within cells and can be released via exosomes, facilitating communication between cells. Studies have demonstrated that exosomal circRNAs can alter the tumor microenvironment and modulate immune responses by influencing the functions of T cells, natural killer (NK) cells, and macrophages, thereby enabling tumors to evade the immune system. Moreover, exosomal circRNAs show potential as diagnostic biomarkers and therapeutic targets for cancer. This review summarizes the regulatory roles of exosomal circRNAs in immune cells and their potential applications in cancer progression and treatment, highlighting their promise in improving cancer immunotherapy. Future research should concentrate on understanding the mechanisms of key exosomal circRNAs and developing targeted immunotherapy methods.
Collapse
Affiliation(s)
- Cunming Lv
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Jinhao Chen
- College of Basic Medical Sciences, China Three Gorges University, Yichang, China
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China
| | - Yuxiang Wang
- College of Electrical Engineering and New Energy, China Three Gorges University, Yichang, China
| | - Yichen Lin
- Department of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
3
|
Li R, Zhou C, Ye K, Chen H, Peng M. Identification of genes involved in energy metabolism in preeclampsia and discovery of early biomarkers. Front Immunol 2025; 16:1496046. [PMID: 39967661 PMCID: PMC11832505 DOI: 10.3389/fimmu.2025.1496046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
Background Preeclampsia is a complex pregnancy condition marked by hypertension and organ dysfunction, posing significant risks to maternal and fetal health. This study investigates the role of energy metabolism-associated genes in preeclampsia development and identifies potential early diagnostic biomarkers. Methods Preeclampsia datasets from Gene Expression Omnibus were analyzed for batch correction, normalization, and differential expression. Enrichment analyses using gene ontology, Kyoto Encyclopedia of Genes and Genomes, and gene set enrichment were performed. Protein-protein interaction networks were constructed to identify key genes, and regulatory networks involving transcription factors, miRNAs, and RNA-binding proteins were established. Differential expression was validated with receiver operating characteristic curve analyses, and immune infiltration was assessed. Results Six energy metabolism-related genes were identified. Enrichment analyses revealed their involvement in glycolysis, gluconeogenesis, lipid transport, bone remodeling, and glucagon secretion. Key differentially expressed genes included CRH(Corticotropin-Releasing Hormone), LEP(Leptin), PDK4(Pyruvate Dehydrogenase Kinase Isozyme 4), SPP1(Secreted Phosphoprotein 1), and SST(Somatostatin). PDK4 exhibited moderate accuracy in receiver operating characteristic analysis. Immune infiltration analysis indicated significant differences between preeclampsia and control samples. qRT-PCR confirmed LEP and CRH increased, while SPP1 expression in preeclampsia samples. Conclusion Dysregulated energy metabolism-related genes may contribute to preeclampsia through metabolic and immune changes. Identifying these genes aids in understanding preeclampsia's molecular basis and early diagnosis. Future studies should validate these markers in larger cohorts and explore targeted treatments.
Collapse
Affiliation(s)
| | | | | | | | - Mengjia Peng
- Department of Gynecology and Obstetrics, The Third Affiliated Hospital of Wenzhou
Medical University, Rui’an, China
| |
Collapse
|
4
|
Milovanovic B, Markovic N, Petrovic M, Zugic V, Ostojic M, Rankovic-Nicic L, Bojic M. Assessment of Autonomic Nervous System Function in Patients with Chronic Fatigue Syndrome and Post-COVID-19 Syndrome Presenting with Recurrent Syncope. J Clin Med 2025; 14:811. [PMID: 39941481 PMCID: PMC11818862 DOI: 10.3390/jcm14030811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/18/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Chronic fatigue syndrome and post-COVID-19 syndrome are associated with dysfunction of the autonomic nervous system, which may manifest as syncope and orthostatic intolerance. This study aimed to compare autonomic nervous system function in patients with chronic fatigue syndrome of unknown etiology and those with chronic fatigue syndrome secondary to post-COVID-19 syndrome using multiple diagnostic modalities, and to assess the prevalence and characteristics of syncope in these populations. Methods: This cross-sectional study included 440 patients examined at the Neurocardiological Laboratory of the Institute for Cardiovascular Diseases "Dedinje". Patients were divided into three groups: chronic fatigue syndrome of unknown etiology (Group 1, n = 210), chronic fatigue syndrome secondary to post-COVID-19 syndrome (Group 2, n = 137), and healthy controls (Group 3, n = 91). Diagnostic modalities included cardiovascular reflex tests, the head-up tilt test, beat-to-beat analysis, 24 h Holter electrocardiogram monitoring, and 24 h ambulatory blood pressure monitoring. Statistical analyses were performed using analysis of variance, Tukey's honestly significant difference test, and the Mann-Whitney U test. Results: Both chronic fatigue syndrome groups demonstrated significant autonomic nervous system dysfunction compared to healthy controls (p < 0.05), including reduced baroreceptor sensitivity and impaired heart rate variability parameters. Syncope prevalence was high in both chronic fatigue syndrome groups, with extreme blood pressure variability observed in 45-47% of patients during the head-up tilt test. Patients with post-COVID-19 chronic fatigue syndrome exhibited greater blood pressure increases during the head-up tilt test than those with chronic fatigue syndrome of unknown etiology (p < 0.05). Conclusions: Patients with chronic fatigue syndrome, irrespective of etiology, exhibit significant autonomic nervous system dysfunction and a high prevalence of syncope. Post-COVID-19 chronic fatigue syndrome demonstrates distinct hemodynamic patterns, suggesting unique pathophysiological mechanisms that warrant further investigation.
Collapse
Affiliation(s)
- Branislav Milovanovic
- Institute for Cardiovascular Diseases “Dedinje”, 11000 Belgrade, Serbia; (B.M.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Nikola Markovic
- Institute for Cardiovascular Diseases “Dedinje”, 11000 Belgrade, Serbia; (B.M.)
| | - Masa Petrovic
- Institute for Cardiovascular Diseases “Dedinje”, 11000 Belgrade, Serbia; (B.M.)
| | - Vasko Zugic
- Institute for Cardiovascular Diseases “Dedinje”, 11000 Belgrade, Serbia; (B.M.)
| | - Milijana Ostojic
- Institute for Cardiovascular Diseases “Dedinje”, 11000 Belgrade, Serbia; (B.M.)
| | | | - Milovan Bojic
- Institute for Cardiovascular Diseases “Dedinje”, 11000 Belgrade, Serbia; (B.M.)
| |
Collapse
|
5
|
Warrayat A, Ali A, Waked J, Tocci D, Speth RC. Assessment of the therapeutic potential of salubrinal for ME/CFS and long-COVID. Trends Mol Med 2024:S1471-4914(24)00268-5. [PMID: 39438198 DOI: 10.1016/j.molmed.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024]
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a chronic debilitating condition with no cure that shares commonality with long-COVID. This review examines current understanding of long-COVID symptoms, characteristics of the affected population, the connection with ME/CFS, and the potential for salubrinal, an agent known for its influence on cellular stress pathways, to mitigate these disorders It also describes the historical development and mechanism of action of salubrinal, to mitigate endoplasmic reticulum (ER)/cellular stress responses, that could potentially contribute to symptom improvement in both ME/CFS and long-COVID patients. Further research and clinical trials are warranted to advance our understanding of the potential role of salubrinal in improving the quality of life for individuals with long-COVID-related ME/CFS symptoms as well as ME/CFS patients.
Collapse
Affiliation(s)
- Aseel Warrayat
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Ayah Ali
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Joulin Waked
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Darcy Tocci
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Robert C Speth
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, DC 20007, USA.
| |
Collapse
|
6
|
Baraniuk JN, Eaton-Fitch N, Marshall-Gradisnik S. Meta-analysis of natural killer cell cytotoxicity in myalgic encephalomyelitis/chronic fatigue syndrome. Front Immunol 2024; 15:1440643. [PMID: 39483457 PMCID: PMC11524851 DOI: 10.3389/fimmu.2024.1440643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/09/2024] [Indexed: 11/03/2024] Open
Abstract
Reduced natural killer (NK) cell cytotoxicity is the most consistent immune finding in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). Meta-analysis of the published literature determined the effect size of the decrement in ME/CFS. Databases were screened for papers comparing NK cell cytotoxicity in ME/CFS and healthy controls. A total of 28 papers and 55 effector:target cell ratio (E:T) data points were collected. Cytotoxicity in ME/CFS was significantly reduced to about half of healthy control levels, with an overall Hedges' g of 0.96 (0.75-1.18). Heterogeneity was high but was explained by the range of E:T ratios, different methods, and potential outliers. The outcomes confirm reproducible NK cell dysfunction in ME/CFS and will guide studies using the NK cell model system for pathomechanistic investigations. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024542140.
Collapse
Affiliation(s)
- James N. Baraniuk
- Department of Medicine, Georgetown University, Washington, DC, United States
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Natalie Eaton-Fitch
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| | - Sonya Marshall-Gradisnik
- National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
7
|
Sun Y, Zhang Z, Qiao Q, Zou Y, Wang L, Wang T, Lou B, Li G, Xu M, Wang Y, Zhang Z, Hou X, Chen L, Zhao R. Immunometabolic changes and potential biomarkers in CFS peripheral immune cells revealed by single-cell RNA sequencing. J Transl Med 2024; 22:925. [PMID: 39394558 PMCID: PMC11468054 DOI: 10.1186/s12967-024-05710-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/27/2024] [Indexed: 10/13/2024] Open
Abstract
The pathogenesis of Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) remains unclear, though increasing evidence suggests inflammatory processes play key roles. In this study, single-cell RNA sequencing (scRNA-seq) of peripheral blood mononuclear cells (PBMCs) was used to decipher the immunometabolic profile in 4 ME/CFS patients and 4 heathy controls. We analyzed changes in the composition of major PBMC subpopulations and observed an increased frequency of total T cells and a significant reduction in NKs, monocytes, cDCs and pDCs. Further investigation revealed even more complex changes in the proportions of cell subpopulations within each subpopulation. Gene expression patterns revealed upregulated transcription factors related to immune regulation, as well as genes associated with viral infections and neurodegenerative diseases.CD4+ and CD8+ T cells in ME/CFS patients show different differentiation states and altered trajectories, indicating a possible suppression of differentiation. Memory B cells in ME/CFS patients are found early in the pseudotime, indicating a unique subtype specific to ME/CFS, with increased differentiation to plasma cells suggesting B cell overactivity. NK cells in ME/CFS patients exhibit reduced cytotoxicity and impaired responses, with reduced expression of perforin and CD107a upon stimulation. Pseudotime analysis showed abnormal development of adaptive immune cells and an enhanced cell-cell communication network converging on monocytes in particular. Our analysis also identified the estrogen-related receptor alpha (ESRRA)-APP-CD74 signaling pathway as a potential biomarker for ME/CFS in peripheral blood. In addition, data from the GSE214284 database confirmed higher ESRRA expression in the monocyte cell types of male ME/CFS patients. These results suggest a link between immune and neurological symptoms. The results support a disease model of immune dysfunction ranging from autoimmunity to immunodeficiency and point to amyloidotic neurodegenerative signaling pathways in the pathogenesis of ME/CFS. While the study provides important insights, limitations include the modest sample size and the evaluation of peripheral blood only. These findings highlight potential targets for diagnostic biomarkers and therapeutic interventions. Further research is needed to validate these biomarkers and explore their clinical applications in managing ME/CFS.
Collapse
Affiliation(s)
- Yujing Sun
- Department of Endocrinology and Metabolism, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250012, China
- Shandong Provincial Key Laboratory of Spatiotemporal Regulation and Precision Intervention in Endocrine and Metabolic Diseases;Shandong Provincial Engineering Research Center for Advanced Technologies in Prevention and Treatment of Chromic Metabolic Diseases, Jinan, 250012, Shandong Province, China
| | - Zhenhua Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China
| | - Qincheng Qiao
- Department of Endocrinology and Metabolism, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Ying Zou
- Department of Endocrinology and Metabolism, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Lina Wang
- Department of Endocrinology and Metabolism, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Tixiao Wang
- Department of Endocrinology and Metabolism, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Bo Lou
- Department of Neurology, the Third People's Hospital of Liaocheng, Liaocheng, 252000, Shandong Province, China
| | - Guosheng Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Miao Xu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Yanxiang Wang
- Jinan AXZE Medical Test Laboratory, Jinan, 250012, Shandong Province, China
| | - Zhenhong Zhang
- Jinan AXZE Medical Test Laboratory, Jinan, 250012, Shandong Province, China
| | - Xinguo Hou
- Department of Endocrinology and Metabolism, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250012, China
- Shandong Provincial Key Laboratory of Spatiotemporal Regulation and Precision Intervention in Endocrine and Metabolic Diseases;Shandong Provincial Engineering Research Center for Advanced Technologies in Prevention and Treatment of Chromic Metabolic Diseases, Jinan, 250012, Shandong Province, China
| | - Li Chen
- Department of Endocrinology and Metabolism, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250012, China
- Shandong Provincial Key Laboratory of Spatiotemporal Regulation and Precision Intervention in Endocrine and Metabolic Diseases;Shandong Provincial Engineering Research Center for Advanced Technologies in Prevention and Treatment of Chromic Metabolic Diseases, Jinan, 250012, Shandong Province, China
| | - Ruxing Zhao
- Department of Endocrinology and Metabolism, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250012, China.
- Shandong Provincial Key Laboratory of Spatiotemporal Regulation and Precision Intervention in Endocrine and Metabolic Diseases;Shandong Provincial Engineering Research Center for Advanced Technologies in Prevention and Treatment of Chromic Metabolic Diseases, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
8
|
Giloteaux L, Glass KA, Germain A, Franconi CJ, Zhang S, Hanson MR. Dysregulation of extracellular vesicle protein cargo in female myalgic encephalomyelitis/chronic fatigue syndrome cases and sedentary controls in response to maximal exercise. J Extracell Vesicles 2024; 13:e12403. [PMID: 38173127 PMCID: PMC10764978 DOI: 10.1002/jev2.12403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/27/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024] Open
Abstract
In healthy individuals, physical exercise improves cardiovascular health and muscle strength, alleviates fatigue and reduces the risk of chronic diseases. Although exercise is suggested as a lifestyle intervention to manage various chronic illnesses, it negatively affects people with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), who suffer from exercise intolerance. We hypothesized that altered extracellular vesicle (EV) signalling in ME/CFS patients after an exercise challenge may contribute to their prolonged and exacerbated negative response to exertion (post-exertional malaise). EVs were isolated by size exclusion chromatography from the plasma of 18 female ME/CFS patients and 17 age- and BMI-matched female sedentary controls at three time points: before, 15 min, and 24 h after a maximal cardiopulmonary exercise test. EVs were characterized using nanoparticle tracking analysis and their protein cargo was quantified using Tandem Mass Tag-based (TMT) proteomics. The results show that exercise affects the EV proteome in ME/CFS patients differently than in healthy individuals and that changes in EV proteins after exercise are strongly correlated with symptom severity in ME/CFS. Differentially abundant proteins in ME/CFS patients versus controls were involved in many pathways and systems, including coagulation processes, muscle contraction (both smooth and skeletal muscle), cytoskeletal proteins, the immune system and brain signalling.
Collapse
Affiliation(s)
- Ludovic Giloteaux
- Department of Molecular Biology and GeneticsCornell UniversityIthacaNew YorkUSA
| | - Katherine A. Glass
- Department of Molecular Biology and GeneticsCornell UniversityIthacaNew YorkUSA
| | - Arnaud Germain
- Department of Molecular Biology and GeneticsCornell UniversityIthacaNew YorkUSA
| | - Carl J. Franconi
- Department of Molecular Biology and GeneticsCornell UniversityIthacaNew YorkUSA
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Institute of BiotechnologyCornell UniversityIthacaNew YorkUSA
| | - Maureen R. Hanson
- Department of Molecular Biology and GeneticsCornell UniversityIthacaNew YorkUSA
| |
Collapse
|
9
|
Hebebrand J, Antel J, von Piechowski L, Kiewert C, Stüve B, Gradl-Dietsch G. Case report: Rapid improvements of anorexia nervosa and probable myalgic encephalomyelitis/chronic fatigue syndrome upon metreleptin treatment during two dosing episodes. Front Psychiatry 2023; 14:1267495. [PMID: 38025476 PMCID: PMC10666640 DOI: 10.3389/fpsyt.2023.1267495] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
A comorbidity of anorexia nervosa (AN) and myalgic encephalomyelitis (ME/CSF) is uncommon. A 17 years-old male adolescent with possible onset of ME/CFS after an Epstein Barr Virus infection (EBV) and later onset of AN during a second period of weight loss was twice treated off-label with metreleptin for 15 and 11 days, respectively. As in previous cases, eating disorder specific cognitions and mood improved. Interestingly, fatigue and post-exertional muscle pain (P-EMP) improved, too. We discuss potential mechanisms. Treatment with metreleptin may prove beneficial in AN and in ME/CSF associated with substantial weight loss.
Collapse
Affiliation(s)
- Johannes Hebebrand
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jochen Antel
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Linda von Piechowski
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Münster, University of Münster, Münster, Germany
| | - Cordula Kiewert
- Division of Pediatric Endocrinology and Diabetology, Department of Pediatrics II, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Burkhard Stüve
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Neuropediatrics, DRK Children’s Hospital, Siegen, Germany
| | - Gertraud Gradl-Dietsch
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|