1
|
Kronenberger D, Zimmers T, Ralston R, Runco D. Circulating Growth Differentiation Factor 15 (GDF15) in Paediatric Disease: A Systematic Review. J Cachexia Sarcopenia Muscle 2025; 16:e13712. [PMID: 40019842 PMCID: PMC11870081 DOI: 10.1002/jcsm.13712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Growth Differentiation Factor 15 (GDF15), a nonspecific inflammatory marker and member of the TGF-β superfamily, has a well-established role in both inflammation and metabolic modulation, but lacks a comprehensive paediatric literature review. In several adult disease states, including cancer cachexia and pregnancy, circulation and expression of GDF15 has been of clinical and scientific interest, but little published paediatric data exists. As such, we aim to summarize existing paediatric studies. METHODS This review follows the PRISMA-ScR guidelines for reporting and aims to summarize existing paediatric studies including GDF15, describe disease entities in which GDF15 has been investigated including existing reference ranges, and identify literature gaps to present future clinical and research direction. Our search strategy queried Ovid MEDLINE, Ovid Embase, Cochrane Library and Scopus databases to find original scientific articles measuring GDF15 from birth through children up to age 18. Data relating to study participant demographic and disease pathology, GDF15 measurement methods and clinical outcomes of interest were extracted. RESULTS Sixty-two studies were included, classified as cardiac, endocrine, mitochondrial, hematologic, neonatal, oncologic, infectious, rheumatologic, renal, neurologic or healthy. While several entities demonstrated elevated GDF15, the highest median GDF15 levels were observed in cardiac arrest 7089 pg/mL (interquartile range 3805-13 306) and mitochondrial diseases 4640 pg/mL (1896-14 064). In certain conditions, including cardiac stress, polycystic ovarian syndrome (PCOS), Kawasaki Disease (KD) and certain mitochondrial myopathies GDF15 can normalize with disease treatment or resolution. Of healthy children studied, GDF15 levels were highest in healthy neonates and followed a predictable pattern, decreasing over time. Mean and standard deviation values of GDF15 in healthy children were 343.8 ± 221.0 pg/mL, with a range of 90-1134 pg/mL for study averages. CONCLUSIONS Circulating GDF15 has been studied in a variety of paediatric diseases. However, variable evaluated outcome measures and GDF15 measurement methodologies prevent generalizability and direct comparison of these published studies. Validating normal GDF15 levels in children with standardized and reproducible methodology will help clarify GDF15's utility as a diagnostic marker of disease, a necessary step to elucidate clinical implications of GDF15 over expression and its potential as a therapeutic target.
Collapse
Affiliation(s)
| | - Teresa A. Zimmers
- Department of Cell, Developmental, and Cancer Biology, Knight Cancer InstituteOregon Health & Science UniversityPortlandOregonUSA
| | - Rick K. Ralston
- Ruth Lilly Medical LibraryIndiana University School of MedicineIndianapolisIndianaUSA
| | - Daniel V. Runco
- Division of Hematology/Oncology, Department of Pediatrics, Seattle Children's HospitalUniversity of WashingtonSeattleWashingtonUSA
- Ben Towne Center for Childhood Cancer ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
- Fred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| |
Collapse
|
2
|
Rivero-Segura NA, Cuartas JDR, Garcia-delaTorre P, Sanchez-Garcia S, Ramirez-Aldana R, Gomez-Verjan JC. Insomnia accelerates the epigenetic clocks in older adults. GeroScience 2025:10.1007/s11357-025-01608-7. [PMID: 40100530 DOI: 10.1007/s11357-025-01608-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/08/2025] [Indexed: 03/20/2025] Open
Abstract
Insomnia is a common sleep disorder characterized mainly by poor sleep quality and insufficient sleep duration. It affects a significant proportion of the global population and is correlated with physical and mental consequences such as cognitive decline, anxiety, chronic fatigue, poor concentration, and memory impairment. Interestingly, it is also linked to ageing and age-related diseases (cardiovascular, metabolic, and neurodegenerative). On the other hand, as we age, DNA methylation patterns undergo significant changes. These have been used to develop the so-called epigenetic clocks that estimate the biological age linked to the environment and the risk of diseases. Few studies have evaluated the association between insomnia and epigenetic clocks, providing insight into the role of insomnia in ageing acceleration. Therefore, in the present study, we carried out an epigenetic analysis by using Illumina EPICv.2 array on 63 older adults (> 60 years old, n = 33 with insomnia vs. n = 30 control) to evaluate the relation between insomnia and epigenetic ages (HorvathAGE, HannumAGE, PhenoAGE, SkinBloodClock, GrimAGE, DunedinPACE, DNAmTL). As a result, we found an increased acceleration and correlation between GrimAGE and SkinBloodClock and a significant reduction in the DNAmTL in individuals with insomnia. An EWAS analysis showed a global pattern of hypomethylation and an enrichment of several proteostasis and oxidative pathways. In conclusion, our results suggest that insomnia increases GrimAGE and SkinBloodClock acceleration and may be participating in telomere shortening. Additionally, changes in DNA methylation patterns induced by insomnia impact proteostasis and oxidative stress.
Collapse
Affiliation(s)
| | | | - Paola Garcia-delaTorre
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, 06720, Mexico City, Mexico
| | - Sergio Sanchez-Garcia
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, 06720, Mexico City, Mexico
| | - Ricardo Ramirez-Aldana
- Escuela Superior de Ingeniería y Tecnología, Universidad Internacional de la Rioja, Logroño, Spain
- Departamento de Matemáticas, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Juan Carlos Gomez-Verjan
- Dirección de Investigación, Instituto Nacional de Geriatría (INGER), 10200, Mexico City, Mexico.
| |
Collapse
|
3
|
Wimalanathan T, Paus MF, Brox Skranes J, Berge T, Tveit A, Røsjø H, Omland T, Lyngbakken MN, Lagethon Heck S. Associations between Growth Differentiation Factor 15, Cardiac Troponin T, and N-terminal pro-B-type Natriuretic Peptide, and Future Myocardial Fibrosis Assessed by Cardiac Magnetic Resonance Imaging: Data from the Akershus Cardiac Examination 1950 Study. J Appl Lab Med 2025; 10:392-405. [PMID: 39707823 DOI: 10.1093/jalm/jfae145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/21/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Myocardial fibrosis is associated with a poor outcome for patients with cardiovascular disease (CVD). Growth differentiation factor 15 (GDF-15) concentrations predict the risk of death in patients with CVD, but the underlying pathophysiological mechanisms are poorly understood. We aimed to assess the associations between biomarkers of cellular stress and inflammation (GDF-15), cardiac injury (cardiac troponin T [cTnT]), and stretch (N-terminal pro-B-type natriuretic peptide [NT-proBNP]), and subsequent focal and diffuse myocardial fibrosis assessed by cardiac magnetic resonance (CMR) imaging. METHODS We measured GDF-15, cTnT, and NT-proBNP in 200 study participants without known coronary artery disease or renal dysfunction from the population-based Akershus Cardiac Examination 1950 Study at baseline in 2012 to 2015. Focal myocardial scars and diffuse fibrosis were assessed by late gadolinium enhancement imaging and septal extracellular volume fraction (ECV) by CMR 4 to 7 years later. The relationships between cardiac biomarkers and CMR parameters were assessed by logistic regression analysis adjusted for common cardiovascular risk factors. RESULTS The median age was 63.9 (interquartile range 63.4-64.5) years and 49% were women. GDF-15 (adjusted odds ratio [aOR] 4.40, 95% CI 1.09-17.72) and cTnT (aOR 1.59, 95% CI 1.01-2.50) were associated with nonischemic scars in the fully adjusted model. cTnT (aOR 2.45, 95% CI 1.41-4.25) and NT-proBNP (aOR 3.12, 95% CI 1.55-6.28) were associated with ischemic scars. None of the biomarkers were significantly associated with elevated ECV. CONCLUSIONS In a general population cohort, GDF-15, an emerging biomarker of cellular stress and inflammation, associates with nonischemic scars. Biomarkers of myocardial injury and stretch associate with ischemic scars, while no biomarker was associated with diffuse fibrosis as assessed by CMR.
Collapse
Affiliation(s)
- Thakshani Wimalanathan
- Department of Diagnostic Imaging, Division of Diagnostics and Technology, Akershus University Hospital, Lørenskog, Norway
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Michael Fredrik Paus
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Julia Brox Skranes
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trygve Berge
- Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Arnljot Tveit
- Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Helge Røsjø
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Akershus Clinical Research Center (ACR), Division of Research and Innovation, Akershus University Hospital, Lørenskog, Norway
| | - Torbjørn Omland
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - M N Lyngbakken
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Siri Lagethon Heck
- Department of Diagnostic Imaging, Division of Diagnostics and Technology, Akershus University Hospital, Lørenskog, Norway
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
4
|
Wittczak A, Mazurek-Kula A, Banach M, Piotrowski G, Bielecka-Dabrowa A. Blood Biomarkers as a Non-Invasive Method for the Assessment of the State of the Fontan Circulation. J Clin Med 2025; 14:496. [PMID: 39860501 PMCID: PMC11765985 DOI: 10.3390/jcm14020496] [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: 12/22/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
The Fontan operation has become the primary palliative treatment for patients with a functionally univentricular heart. The population of patients with Fontan circulation is constantly growing and aging. As the number of Fontan patients surviving into adulthood increases, there is a clear need for research on how best to follow these patients and manage their complications. Monitoring blood biomarkers is a promising method for the non-invasive assessment of the Fontan circulation. In this article, we provide a comprehensive review of the available evidence on this topic. The following biomarkers were included: natriuretic peptides, red blood cell distribution width (RDW), cystatin C, high-sensitivity C-reactive protein, vitamin D, parathyroid hormone, von Willebrand factor, carbohydrate antigen 125, lipoproteins, hepatocyte growth factor, troponins, ST2 protein, galectin-3, adrenomedullin, endothelin-1, components of the renin-angiotensin-aldosterone system, norepinephrine, interleukin 6, tumor necrosis factor α, and uric acid. We did not find strong enough data to propose evidence-based recommendations. Nevertheless, significantly elevated levels of brain natriuretic peptide (BNP)/N-terminal prohormone of BNP (NT-proBNP) are most likely associated with the failure of the Fontan circulation. The use of the RDW is also promising. Several biomarkers appear to be useful in certain clinical presentations. Certainly, robust longitudinal, preferably multicenter, prospective studies are needed to determine the sensitivity, specificity, evidence-based cut-off values and overall predictive value of different biomarkers in monitoring Fontan physiology.
Collapse
Affiliation(s)
- Andrzej Wittczak
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, 90-419 Lodz, Poland
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland
| | - Anna Mazurek-Kula
- Department of Cardiology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, 90-419 Lodz, Poland
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland
| | - Grzegorz Piotrowski
- Cardiooncology Department, Medical University of Lodz, 90-419 Lodz, Poland
- Cardiology Department, Nicolaus Copernicus Memorial Hospital, 93-513 Lodz, Poland
| | - Agata Bielecka-Dabrowa
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, 90-419 Lodz, Poland
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland
| |
Collapse
|
5
|
Buchan E, Harbi MH, Rickard JJS, Thomas M, Goldberg Oppenheimer P. Advanced biomolecular spectroscopic profiling of cardiovascular disease macromolecular markers: SIL-6, IL-9, LpA, ApoB, PCSK9 and NT-ProBNP for rapid in-situ detection and monitoring. Int J Biol Macromol 2025; 284:138115. [PMID: 39608533 DOI: 10.1016/j.ijbiomac.2024.138115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Cardiovascular disease (CVD) remains a major global health concern and a leading cause of morbidity and mortality worldwide. Early-diagnosis and prompt medical attention are crucial in managing and reducing overall impact on health-and-wellbeing, necessitating the development of innovative diagnostics, which transcend traditional methodologies. Raman spectroscopy uniquely provides molecular fingerprinting and structural information, offering insights into biochemical composition. Integration of Raman spectroscopy with advanced machine learning is established as a powerful clinical adjunct for point-of-care detection of CVDs. A non-invasive, label-free spectroscopic platform coupled with neural network algorithm, 'SKiNET' has been developed to accurately detect the biomolecular changes within plasma of CVD versus healthy cohorts, enabling rapid diagnosis and longer-term monitoring, where the real-time capabilities provide dynamic assessment of progression, aligning treatment strategies with evolving states. CVD has been detected and classified via SKiNET with 88.6 %-accuracy, 92.9 %-specificity and 85.1 %-sensitivity and with 83.8 %-accuracy. The hybrid RS-SKiNET bio-molecularly specific detection signposted a comprehensive panel of CVD-indicative biomarkers, including SIL-6, IL-9, LpA, ApoB, PCSK9 and NT-ProBNP, offering important insights into disease mechanisms and risk-stratification. This multidimensional technique holds potential for improved patient-and-healthcare management for CVDs, laying the platform toward high-throughput biomolecular profiling of CVD-indicative macromolecular biomarkers, particularly vital for widespread point-of-care diagnostics and monitoring.
Collapse
Affiliation(s)
- Emma Buchan
- School of Chemical Engineering, College of Engineering and Physical Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Maan H Harbi
- Pharmacology and Toxicology Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Jonathan J S Rickard
- Department of Physics, Cavendish Laboratory, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
| | - Mark Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Pola Goldberg Oppenheimer
- School of Chemical Engineering, College of Engineering and Physical Science, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Healthcare Technologies Institute, Institute of Translational Medicine, Mindelsohn Way, Birmingham B15 2TH, UK.
| |
Collapse
|
6
|
Skau E, Wagner P, Leppert J, Ärnlöv J, Hedberg P. Determinants of growth differentiation factor 15 plasma levels in outpatients with peripheral arterial disease. Ups J Med Sci 2024; 129:11001. [PMID: 39780955 PMCID: PMC11708457 DOI: 10.48101/ujms.v129.11001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/27/2024] [Accepted: 08/27/2024] [Indexed: 01/11/2025] Open
Abstract
Background Growth differentiation factor 15 (GDF-15) is a robust prognostic biomarker in patients with cardiovascular (CV) disease, and a better understanding of its clinical determinants is desirable. We aimed to study the associations between GDF-15 levels and traditional CV risk factors, indicators of atherosclerotic burden, and cardiac geometry and dysfunction in outpatients with peripheral arterial disease (PAD). Methods An explorative cross-sectional study (Study of Atherosclerosis in Vastmanland, Västerås, Sweden) included 439 outpatients with carotid or lower extremity PAD. The mean age was 70 years (standard deviation [SD] 7), and 59% of the patients were men. Plasma levels of GDF-15 were obtained along with potential determinants, including medical history, biochemical data, echocardiographic measures of cardiac geometry and function, ankle-brachial index (ABI), and carotid ultrasonographic data on intima-media thickness (IMT) and occurrence of carotid stenosis. The relations between GDF-15 concentrations (transformed with the natural logarithm) and the different determinants were evaluated using uni- and multivariable linear regression models. All pre-specified variables were included in the multivariable models. Results The multivariable analysis identified independent relations of GDF-15 with several of the included variables (adjusted R 2 = 0.48). Diabetes (beta coefficient [β] of 0.37, 95% confidence interval [95% CI] 0.25 to 0.50), low-density lipoprotein (LDL) cholesterol (β = -0.22, 95% confidence interval [CI]: -0.34 to -0.09), and physical activity (β = -0.16, 95% CI: -0.25 to -0.06) had the strongest associations. In contrast, no significant independent associations with GDF-15 level were observed for cardiac geometry and function, ABI, IMT, or carotid stenosis. Conclusions Circulating GDF-15 is more strongly associated with traditional CV risk factors, especially diabetes, LDL cholesterol, and physical activity than with specific indicators of atherosclerotic burden or cardiac dysfunction. To better understand the pathophysiological role of GDF-15 and its link to clinical outcomes in patients with PAD, future studies should focus on the metabolic processes involved in atherosclerotic disease.
Collapse
Affiliation(s)
- Emma Skau
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
- Department of Cardiology, Danderyd University Hospital, Stockholm, Sweden
| | - Philippe Wagner
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
| | - Jerzy Leppert
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
| | - Johan Ärnlöv
- School of Health and Social Studies, Dalarna University, Falun, Sweden
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
| | - Pär Hedberg
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
- Department of Clinical Physiology, Västmanland County Hospital, Västerås, Sweden
| |
Collapse
|
7
|
Torrens-Mas M, Navas-Enamorado C, Galmes-Panades A, Masmiquel L, Sanchez-Polo A, Capo X, Gonzalez-Freire M. GDF-15 as a proxy for epigenetic aging: associations with biological age markers, and physical function. Biogerontology 2024; 26:22. [PMID: 39644331 PMCID: PMC11625061 DOI: 10.1007/s10522-024-10165-z] [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: 10/17/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Growth differentiation factor 15 (GDF-15) has emerged as a significant biomarker of aging, linked to various physiological and pathological processes. This study investigates circulating GDF-15 levels in a cohort of healthy individuals from the Balearic Islands, exploring its associations with biological age markers, including multiple DNA methylation (DNAm) clocks, physical performance, and other age-related biomarkers. Seventy-two participants were assessed for general health, body composition, and physical function, with GDF-15 levels quantified using ELISA. Our results indicate that GDF-15 levels significantly increase with age, particularly in individuals over 60. Strong positive correlations were observed between GDF-15 levels and DNAm GrimAge, DNAm PhenoAge, Hannum, and Zhang clocks, suggesting that GDF-15 could serve as a proxy for epigenetic aging. Additionally, GDF-15 levels were linked to markers of impaired glycemic control, systemic inflammation, and physical decline, including decreased lung function and grip strength, especially in men. These findings highlight the use of GDF-15 as a biomarker for aging and age-related functional decline. Given that GDF-15 is easier to measure than DNA methylation, it has the potential to be more readily implemented in clinical settings for broader health assessment and management.
Collapse
Affiliation(s)
- Margalida Torrens-Mas
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d´Investigació en Ciències de La Salut (IUNICS), University of the Balearic Islands, 07122, Palma, Spain
| | - Cayetano Navas-Enamorado
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
| | - Aina Galmes-Panades
- Physical Activity and Sport Sciences Research Group (GICAFE), Institute for Educational Research and Innovation (IRIE), University of the Balearic Islands, 07122, Palma, Spain
- Consorcio CIBER, M.P. Fisiopatología de La Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Luis Masmiquel
- Vascular and Metabolic Pathologies Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
| | - Andrés Sanchez-Polo
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain
| | - Xavier Capo
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain.
- Research Group in Community Nutrition and Oxidative Stress, University of the Balearic Islands-Institut Universitari d´Investigació en Ciències de La Salut (IUNICS), 07122, Palma, Spain.
| | - Marta Gonzalez-Freire
- Translational Research in Aging and Longevity (TRIAL) Group, Health Research Institute of the Balearic Islands (IdISBa), 07120, Palma, Spain.
| |
Collapse
|
8
|
Lorenz EC, Smith BH, Liang Y, Park WD, Bentall AJ, Dhala AF, Waterman AD, Kennedy CC, Hickson LJ, Rule AD, Cheville AL, LeBrasseur NK, Stegall MD. Increased Pretransplant Inflammatory Biomarkers Predict Death With Function After Kidney Transplantation. Transplantation 2024; 108:2434-2445. [PMID: 38913783 PMCID: PMC11666810 DOI: 10.1097/tp.0000000000005103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
BACKGROUND Chronic systemic inflammation is associated with mortality in patients with chronic kidney disease, cardiovascular disease, and diabetes. The goal of this study was to examine the relationship between pretransplant inflammatory biomarkers (growth differentiation factor-15 [GDF-15], interleukin-6 [IL-6], soluble tumor necrosis factor receptor-1, monokine induced by gamma interferon/chemokine [C-X-C motif] ligand 9 [MIG/CXCL9], monocyte chemoattractant protein-1, soluble FAS, tumor necrosis factor-α, interleukin-15, and interleukin-1β) and death with function (DWF) after kidney transplantation (KT). METHODS We retrospectively measured inflammatory biomarker levels in serum collected up to 1 y before KT (time from blood draw to KT was 130 ± 110 d) in recipients transplanted between January 2006 and December 2018. Kaplan-Meier estimation, Cox regression, and Gradient Boosting Machine modeling were used to examine the relationship between inflammatory biomarkers and DWF. RESULTS Our cohort consisted of 1595 KT recipients, of whom 62.9% were male and 83.2% were non-Hispanic White. Over a mean follow-up of 7.4 ± 3.9 y, 21.2% of patients (n = 338) experienced DWF. Patients with the highest quartile levels of GDF-15 (>4766 pg/mL), IL-6 (>6.11 pg/mL), and MIG/CXCL9 (> 5835 pg/mL) had increased rates of DWF, and each predicted mortality independently of the others. When adjusted for clinical factors (age, diabetes, etc), the highest quartile levels of GDF-15 and IL-6 remained independently associated with DWF. Adding inflammatory markers to a clinical Cox model improved the C-statistic for DWF from 0.727 to 0.762 using a Gradient Boosting Machine modeling approach. CONCLUSIONS These findings suggest that pre-KT serum concentrations of GDF-15, IL-6, and MIG/CXCL9 may help to risk stratify and manage patients undergoing KT and suggests that chronic inflammation may play a role in mortality in KT recipients.
Collapse
Affiliation(s)
| | - Byron H Smith
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Yun Liang
- Department of Surgery, Mayo Clinic, Rochester, MN
| | | | - Andrew J Bentall
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Atiya F Dhala
- Department of Surgery, Houston Methodist Hospital, Houston, TX
| | - Amy D Waterman
- Department of Surgery, Houston Methodist Hospital, Houston, TX
| | - Cassie C Kennedy
- Division of Pulmonary, Critical Care, and Sleep Medicine, Mayo Clinic, Rochester, MN
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, FL
| | - Andrew D Rule
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Andrea L Cheville
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Nathan K LeBrasseur
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
9
|
Mulcahy MC, El Habbal N, Redd JR, Sun H, Gregg BE, Bridges D. GDF15 Knockout Does Not Substantially Impact Perinatal Body Weight or Neonatal Outcomes in Mice. Endocrinology 2024; 165:bqae143. [PMID: 39445824 PMCID: PMC11577612 DOI: 10.1210/endocr/bqae143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/08/2024] [Accepted: 10/22/2024] [Indexed: 10/25/2024]
Abstract
Growth differentiation factor-15 (GDF15) increases in circulation during pregnancy and has been implicated in food intake, weight loss, complications of pregnancy, and metabolic illness. We used a Gdf15 knockout mouse model (Gdf15-/-) to assess the role of GDF15 in body weight regulation and food intake during pregnancy. We found that Gdf15-/- dams consumed a similar amount of food and gained comparable weight during the course of pregnancy compared with Gdf15+/+ dams. Insulin sensitivity on gestational day 16.5 was also similar between genotypes. In the postnatal period, litter size and survival rates were similar between genotypes. There was a modest reduction in birth weight of Gdf15-/- pups, but this difference was no longer evident from postnatal day 3.5 to 14.5. We observed no detectable differences in milk volume production or milk fat percentage. These data suggest that GDF15 is dispensable for changes in food intake, and body weight as well as insulin sensitivity during pregnancy in a mouse model.
Collapse
Affiliation(s)
- Molly C Mulcahy
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Noura El Habbal
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- School of Health Professions, New York Institute of Technology, Old Westbury, New York, NY 11568, USA
| | - JeAnna R Redd
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Haijing Sun
- Department of Pediatric Endocrinology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Brigid E Gregg
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Department of Pediatric Endocrinology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Dave Bridges
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Varhegyi V, Modos A, Trager D, Gerszi D, Horvath EM, Sipos M, Acs N, Molnar MJ, Varbiro S, Gal A. GDF-15 and mtDNA Deletions Are Useful Biomarkers of Mitochondrial Dysfunction in Insulin Resistance and PCOS. Int J Mol Sci 2024; 25:10916. [PMID: 39456699 PMCID: PMC11507876 DOI: 10.3390/ijms252010916] [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: 09/02/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
There is no literature available about the growth differentiation factor-15 (GDF-15) biomarker in combination with mitochondrial DNA (mtDNA) deletions in insulin resistance (IR), and polycystic ovary syndrome (PCOS); however, it would be useful to achieve optimal metabolic status and improve pregnancy success. In this study, the role of GDF-15 and mtDNA deletions as biomarkers in the pathogenesis of IR and PCOS was investigated. In our study, 81 female patients who were treated for IR and/or PCOS and 41 healthy controls were included. GDF-15 levels in patients showed a marked increase compared to controls. Elevated GDF-15 levels were found in 12 patients; all of them had a BMI > 25 kg/m2, which is associated with reactive hyperinsulinemia. The presence of mitochondrial dysfunction was mainly observed in the IR-only subgroup. The increase in plasma levels of GDF-15 and the prevalence of mtDNA deletions is directly proportional to body mass index. The more marked metabolic abnormalities required more intensive drug therapy with a parallel increase in plasma GDF-15 levels. Elevated levels of GDF-15 and the presence of mitochondrial DNA deletions may be a consequence of carbohydrate metabolism disorders in patients and thus a predictor of the process of accelerated aging.
Collapse
Affiliation(s)
- Vera Varhegyi
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, 1085 Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, 1085 Budapest, Hungary
| | - Anna Modos
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, 1085 Budapest, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, 1085 Budapest, Hungary
| | - Domonkos Trager
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, 1085 Budapest, Hungary
| | - Dora Gerszi
- Department of Obstetrics and Gynecology, Semmelweis University, 1085 Budapest, Hungary
| | | | - Miklos Sipos
- Department of Obstetrics and Gynecology, Semmelweis University, 1085 Budapest, Hungary
| | - Nandor Acs
- Department of Obstetrics and Gynecology, Semmelweis University, 1085 Budapest, Hungary
| | - Maria Judit Molnar
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, 1085 Budapest, Hungary
| | - Szabolcs Varbiro
- Department of Obstetrics and Gynecology, Semmelweis University, 1085 Budapest, Hungary
- Department of Obstetrics and Gynecology, Albert Szent-Györgyi Clinical Centre, University of Szeged, 6720 Szeged, Hungary
- Workgroup for Science Management, Doctoral School, Semmelweis University, 1085 Budapest, Hungary
| | - Aniko Gal
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
11
|
Gurtan AM, Khalid S, Koch C, Khan MZ, Lamarche LB, Splawski I, Dolan E, Carrion AM, Zessis R, Clement ME, Chen Z, Lindsley LD, Chiu YH, Streeper RS, Denning DP, Goldfine AB, Doyon B, Abbasi A, Harrow JL, Tsunoyama K, Asaumi M, Kou I, Shuldiner AR, Rodriguez-Flores JL, Rasheed A, Jahanzaib M, Mian MR, Liaqat MB, Raza SS, Sultana R, Jalal A, Saeed MH, Abbas S, Memon FR, Ishaq M, Dominy JE, Saleheen D. Identification and characterization of human GDF15 knockouts. Nat Metab 2024; 6:1913-1921. [PMID: 39327531 DOI: 10.1038/s42255-024-01135-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024]
Abstract
Growth differentiation factor 15 (GDF15) is a secreted protein that regulates food intake, body weight and stress responses in pre-clinical models1. The physiological function of GDF15 in humans remains unclear. Pharmacologically, GDF15 agonism in humans causes nausea without accompanying weight loss2, and GDF15 antagonism is being tested in clinical trials to treat cachexia and anorexia. Human genetics point to a role for GDF15 in hyperemesis gravidarum, but the safety or impact of complete GDF15 loss, particularly during pregnancy, is unknown3-7. Here we show the absence of an overt phenotype in human GDF15 loss-of-function carriers, including stop gains, frameshifts and the fully inactivating missense variant C211G3. These individuals were identified from 75,018 whole-exome/genome-sequenced participants in the Pakistan Genomic Resource8,9 and recall-by-genotype studies with family-based recruitment of variant carrier probands. We describe 8 homozygous ('knockouts') and 227 heterozygous carriers of loss-of-function alleles, including C211G. GDF15 knockouts range in age from 31 to 75 years, are fertile, have multiple children and show no consistent overt phenotypes, including metabolic dysfunction. Our data support the hypothesis that GDF15 is not required for fertility, healthy pregnancy, foetal development or survival into adulthood. These observations support the safety of therapeutics that block GDF15.
Collapse
Affiliation(s)
| | - Shareef Khalid
- Columbia University Irving Medical Center, New York, NY, USA
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | | | | | | | - Igor Splawski
- Biomedical Research at Novartis, Boston, MA, USA
- Yarrow Biotechnology, New York, NY, USA
| | | | | | | | | | - Zhiping Chen
- Biomedical Research at Novartis, Boston, MA, USA
| | | | - Yu-Hsin Chiu
- Biomedical Research at Novartis, Boston, MA, USA
| | | | | | | | - Brian Doyon
- Biomedical Research at Novartis, Boston, MA, USA
- Tango Therapeutics, Boston, MA, USA
| | - Ali Abbasi
- Centre for Genomics Research, Discovery Sciences, AstraZeneca, Cambridge, UK
| | - Jennifer L Harrow
- Centre for Genomics Research, Discovery Sciences, AstraZeneca, Cambridge, UK
| | | | | | - Ikuyo Kou
- Astellas Pharma Inc., Ibaraki, Japan
| | - Alan R Shuldiner
- Regeneron Genetics Center, LLC, Regeneron Pharmaceuticals Inc., New York, NY, USA
| | | | - Asif Rasheed
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | | | | | | | | | | | - Anjum Jalal
- Punjab Institute of Cardiology, Lahore, Pakistan
| | | | - Shahid Abbas
- Faisalabad Institute of Cardiology, Faisalabad, Pakistan
| | | | | | | | - Danish Saleheen
- Columbia University Irving Medical Center, New York, NY, USA.
- Center for Non-Communicable Diseases, Karachi, Pakistan.
| |
Collapse
|
12
|
Yule MS, Brown LR, Skipworth RJE, Laird BJA. Central neural mechanisms of cancer cachexia. Curr Opin Support Palliat Care 2024; 18:138-144. [PMID: 38752576 DOI: 10.1097/spc.0000000000000707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
PURPOSE OF REVIEW Cachexia is a devasting syndrome which impacts a large number of patients with cancer. This review aims to provide a comprehensive overview of the central mechanisms of cancer cachexia. In particular, it focuses on the role of the central nervous system (CNS), the melanocortin system, circulating hormones and molecules which are produced by and act on the CNS and the psychological symptoms of cancer cachexia. RECENT FINDINGS A growing body of evidence suggests that a central mechanism of action underpins this multi-system disorder. Recent research has focused on the role of neuroinflammation that drives the sickness behaviour seen in cancer cachexia, with emphasis on the role of the hypothalamus. Melanocortin receptor antagonists are showing promise in preclinical studies. There are also new pharmacological developments to overcome the short half-life of ghrelin. GDF-15 has been identified as a core target and trials of compounds that interfere with its signalling or its central receptor are underway. SUMMARY Understanding the central mechanisms of cancer cachexia is pivotal for enhancing treatment outcomes in patients. While emerging pharmacological interventions targeting these pathways have shown promise, further research is essential.
Collapse
Affiliation(s)
- Michael S Yule
- St Columba's Hospice
- Edinburgh Cancer Research Centre, University of Edinburgh
| | - Leo R Brown
- Clinical Surgery, University of Edinburgh, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Richard J E Skipworth
- Clinical Surgery, University of Edinburgh, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Barry J A Laird
- St Columba's Hospice
- Edinburgh Cancer Research Centre, University of Edinburgh
| |
Collapse
|
13
|
Zhang J, Zhang J, Wu T, Jin P, Huang C. A method of quantitative chemiluminescence immunoassay for the concentration of Growth differentiation factor-15. MethodsX 2024; 12:102572. [PMID: 38352663 PMCID: PMC10863303 DOI: 10.1016/j.mex.2024.102572] [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: 10/30/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
Growth differentiation factor-15 (GDF-15), a member of the transforming growth factor (TGF-β) superfamily, and is expressed and secreted in response to inflammation, oxidative stress and hypoxia. It has been shown in several studies to be a predictor of heart failure. However, the only kits available on the market are ELISA kits, which are costly and error-prone and are not conducive for clinical use. Here, we developed a chemiluminescence kit which optimized the reaction conditions and the reaction time was reduced to 10 min. We further proved that it can be used to measure GDF-15 in serum or plasma accurately and fastly, and provide additional information for the diagnosis of heart failure disease. Methodological comparison and clinical study verified this method is a reliable, economical and highly automated blood test method.•All necessary steps and the reagents needed are provided.•Reliability of the chemiluminescence immunoassay was verified by analyzing serum GDF-15 levels from different groups.•GDF-15 can provide clinicians with reliable prediction and disease assessment of heart failure.
Collapse
Affiliation(s)
- Ju Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
- Department of Clinical Laboratory Diagnostics, Bengbu Medical College, Bengbu, Anhui, China
| | - Jiajia Zhang
- Department of Clinical Laboratory Diagnostics, Bengbu Medical College, Bengbu, Anhui, China
| | - Ting Wu
- Maccura Biotechnology Co., Ltd, Chengdu, Sichuan, China
| | - Peipei Jin
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, Anhui, China
| | - Chengyi Huang
- Maccura Biotechnology Co., Ltd, Chengdu, Sichuan, China
| |
Collapse
|
14
|
Mulcahy MC, El Habbal N, Redd JR, Sun H, Gregg BE, Bridges D. GDF15 knockout does not substantially impact perinatal body weight or neonatal outcomes in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591359. [PMID: 38746399 PMCID: PMC11092610 DOI: 10.1101/2024.04.30.591359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Growth differentiation factor-15 (GDF15) increases in circulation during pregnancy and has been implicated in food intake, weight loss, complications of pregnancy, and metabolic illness. We used a Gdf15 knockout mouse model (Gdf15-/- ) to assess the role of GDF15 in body weight regulation and food intake during pregnancy. We found that Gdf15-/- dams consumed a similar amount of food and gained comparable weight during the course of pregnancy compared to Gdf15+/+ dams. Insulin sensitivity on gestational day 16.5 was also similar between genotypes. In the postnatal period, litter size, and survival rates were similar between genotypes. There was a modest reduction in birth weight of Gdf15-/- pups, but this difference was no longer evident postnatal day 3.5 to 14.5. We observed no detectable differences in milk volume production or milk fat percentage. These data suggest that GDF15 is dispensable for changes in food intake, and body weight as well as insulin sensitivity during pregnancy in a mouse model.
Collapse
Affiliation(s)
- Molly C. Mulcahy
- University of Michigan School of Public Health, Department of Nutritional Sciences
| | - Noura El Habbal
- University of Michigan School of Public Health, Department of Nutritional Sciences
| | - JeAnna R. Redd
- University of Michigan School of Public Health, Department of Nutritional Sciences
| | - Haijing Sun
- Michigan Medicine, Department of Pediatric Endocrinology
| | - Brigid E. Gregg
- University of Michigan School of Public Health, Department of Nutritional Sciences
- Michigan Medicine, Department of Pediatric Endocrinology
| | - Dave Bridges
- University of Michigan School of Public Health, Department of Nutritional Sciences
| |
Collapse
|
15
|
Pena-Leon V, Perez-Lois R, Villalon M, Folgueira C, Barja-Fernández S, Prida E, Baltar J, Santos F, Fernø J, García-Caballero T, Nogueiras R, Quiñones M, Al-Massadi O, Seoane LM. Gastric GDF15 levels are regulated by age, sex, and nutritional status in rodents and humans. J Endocrinol Invest 2024; 47:1139-1154. [PMID: 37955834 DOI: 10.1007/s40618-023-02232-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/21/2023] [Indexed: 11/14/2023]
Abstract
AIM Growth differentiation factor 15 (GDF15) is a stress response cytokine that has been proposed as a relevant metabolic hormone. Descriptive studies have shown that plasma GDF15 levels are regulated by short term changes in nutritional status, such as fasting, or in obesity. However, few data exist regarding how GDF15 levels are regulated in peripheral tissues. The aim of the present work was to study the variations on gastric levels of GDF15 and its precursor under different physiological conditions, such as short-term changes in nutritional status or overfeeding achieved by HFD. Moreover, we also address the sex- and age-dependent alterations in GDF15 physiology. METHODS The levels of gastric and plasma GDF15 and its precursor were measured in lean and obese mice, rats and humans by western blot, RT-PCR, ELISA, immunohistochemistry and by an in vitro organ culture system. RESULTS Our results show a robust regulation of gastric GDF15 production by fasting in rodents. In obesity an increase in GDF15 secretion from the stomach is reflected with an increase in circulating levels of GDF15 in rats and humans. Moreover, gastric GDF15 levels increase with age in both rats and humans. Finally, gastric GDF15 levels display sexual dimorphism, which could explain the difference in circulating GFD15 levels between males and females, observed in both humans and rodents. CONCLUSIONS Our results provide clear evidence that gastric GDF15 is a critical contributor of circulating GDF15 levels and can explain some of the metabolic effects induced by GDF15.
Collapse
Affiliation(s)
- V Pena-Leon
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - R Perez-Lois
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - M Villalon
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - C Folgueira
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - S Barja-Fernández
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - E Prida
- Translational Endocrinology Group, Endocrinology Section, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (IDIS/CHUS), Santiago de Compostela, Spain
| | - J Baltar
- Servicio de Cirugía General y del Aparato Digestivo, CHUS7SERGAS Santiago de Compostela, Rua R Baltar s/n, 15706, Santiago de Compostela, Spain
| | - F Santos
- Servicio de Cirugía General y del Aparato Digestivo, CHUS7SERGAS Santiago de Compostela, Rua R Baltar s/n, 15706, Santiago de Compostela, Spain
| | - J Fernø
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, 5201, Bergen, Norway
| | - T García-Caballero
- Departamento de Ciencias Morfologicas, Facultad de Medicina, USC, Complejo Hospitalario de Santiago (CHUS/SERGAS), Santiago de Compostela, Spain
| | - R Nogueiras
- Departamento de Fisiología, Instituto de Investigación Sanitaria de Santiago de Compostela, CIMUS, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - M Quiñones
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - O Al-Massadi
- Translational Endocrinology Group, Endocrinology Section, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (IDIS/CHUS), Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
| | - L M Seoane
- Grupo Fisiopatología Endocrina, Área de Endocrinología, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, Spain, Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
| |
Collapse
|
16
|
Nyárády BB, Kiss LZ, Bagyura Z, Merkely B, Dósa E, Láng O, Kőhidai L, Pállinger É. Growth and differentiation factor-15: A link between inflammaging and cardiovascular disease. Biomed Pharmacother 2024; 174:116475. [PMID: 38522236 DOI: 10.1016/j.biopha.2024.116475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024] Open
Abstract
Age-related disorders are closely linked to the accumulation of senescent cells. The senescence-associated secretory phenotype (SASP) sustains and progresses chronic inflammation, which is involved in cellular and tissue dysfunction. SASP-related growth and differentiation factor-15 (GDF-15) is an immunoregulatory cytokine that is coupled to aging and thus may have a regulatory role in the development and maintenance of atherosclerosis, a major cause of cardiovascular disease (CVD). Although the effects of GDF-15 are tissue-specific and dependent on microenvironmental changes such as inflammation, available data suggest that GDF-15 has a significant role in CVD. Thus, GDF-15 is a promising biomarker and potential therapeutic target for atherosclerotic CVD.
Collapse
Affiliation(s)
- Balázs Bence Nyárády
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, Budapest H-1122, Hungary.
| | - Loretta Zsuzsa Kiss
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, Budapest H-1122, Hungary.
| | - Zsolt Bagyura
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, Budapest H-1122, Hungary.
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, Budapest H-1122, Hungary.
| | - Edit Dósa
- Heart and Vascular Center, Semmelweis University, Városmajor utca 68, Budapest H-1122, Hungary.
| | - Orsolya Láng
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, Budapest H-1089, Hungary.
| | - László Kőhidai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, Budapest H-1089, Hungary.
| | - Éva Pállinger
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, Budapest H-1089, Hungary.
| |
Collapse
|
17
|
Welsh P, Kimenai DM, Shah ASV, Gadd DA, Marioni RE, Woodward M, Sudlow CLM, Campbell A, Cleland JGF, Pellicori P, Hayward C, Mills NL, Sattar N. Multiple Cardiac Biomarkers to Improve Prediction of Cardiovascular Events: Findings from the Generation Scotland Scottish Family Health Study. Clin Chem 2024; 70:403-413. [PMID: 38069915 DOI: 10.1093/clinchem/hvad205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/27/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND Many studies have investigated whether single cardiac biomarkers improve cardiovascular risk prediction for primary prevention but whether a combined approach could further improve risk prediction is unclear. We aimed to test a sex-specific, combined cardiac biomarker approach for cardiovascular risk prediction. METHODS In the Generation Scotland Scottish Family Health Study, N-terminal pro-B-type natriuretic peptide (NT-proBNP), growth differentiation factor-15 (GDF-15), cardiac troponin I (cTnI), cardiac troponin T (cTnT), and C-reactive protein (CRP) were measured in stored serum using automated immunoassays. Sex-specific Cox models that included SCORE2 risk factors evaluated addition of single and combined biomarkers for prediction of major adverse cardiovascular events (MACE). Combined biomarker models were compared to a baseline model that included SCORE2 risk factors. RESULTS The study population comprised 18 383 individuals (58.9% women, median age of 48 years [25th-75th percentile, 35-58 years]). During the median follow up of 11.6 (25th-75th percentile, 10.8-13.0) years, MACE occurred in 942 (5.1%) individuals. The greatest increase in discrimination with addition of individual biomarkers to the base model was for women GDF-15 and for men NT-proBNP (change in c-index: + 0.010 for women and +0.005 for men). For women, combined biomarker models that included GDF-15 and NT-proBNP (+0.012) or GDF-15 and cTnI (+0.013), but not CRP or cTnT, further improved discrimination. For men, combined biomarker models that included NT-proBNP and GDF-15 (+0.007), NT-proBNP and cTnI (+0.006), or NT-proBNP and CRP (+0.008), but not cTnT, further improved discrimination. CONCLUSIONS A combined biomarker approach, particularly the use of GDF-15, NT-proBNP and cTnI, further refined cardiovascular risk estimates.
Collapse
Affiliation(s)
- Paul Welsh
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Dorien M Kimenai
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Anoop S V Shah
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Danni A Gadd
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark Woodward
- The George Institute for Global Health, School of Public Health, Imperial College London, London, United Kingdom
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Cathie L M Sudlow
- British Heart Foundation Data Science Centre, Health Data Research UK, London, United Kingdom
- Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - John G F Cleland
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Pierpaolo Pellicori
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicholas L Mills
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
- Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Naveed Sattar
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
18
|
Kiss LZ, Nyárády BB, Pállinger É, Lux Á, Jermendy ÁL, Csobay-Novák C, Soós P, Szelid Z, Láng O, Kőhidai L, Dinya E, Dósa E, Merkely B, Bagyura Z. Association of growth and differentiation factor-15 with coronary artery calcium score and ankle-brachial index in a middle-aged and elderly Caucasian population sample free of manifest cardiovascular disease. GeroScience 2024; 46:1343-1350. [PMID: 37548881 PMCID: PMC10828406 DOI: 10.1007/s11357-023-00899-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/30/2023] [Indexed: 08/08/2023] Open
Abstract
Growth and differentiation factor-15 (GDF-15) is a stress-associated cytokine of the transforming growth factor-β superfamily. The inflammatory and angiogenic effects of GDF-15 in atherosclerosis are controversial, and its correlation with the long asymptomatic phase of the disease is not well understood. Coronary artery calcium score (CACS) and ankle-brachial index (ABI) are sensitive markers of subclinical atherosclerosis. To date, only a few studies have examined the impact of GDF-15 on coronary artery calcification, and the association between GDF-15 and ABI has not been evaluated. Therefore, we aimed to investigate the possible relationship between serum GDF-15 concentrations and CACS and ABI in a Caucasian population sample of middle-aged (35-65 years) and elderly (> 65 years) people. In addition to recording demographic and anthropometric characteristics, atherosclerotic risk factors, and laboratory tests including serum HDL-cholesterol, LDL-cholesterol, hemoglobin A1c (HbA1c), high-sensitivity C-reactive protein, and N-terminal pro-B-type natriuretic peptide (NT-proBNP); GDF-15 level, cardiac computed tomography, and ABI measurements were also performed. A total of 269 asymptomatic individuals (men, n = 125; median age, 61.5 [IQR, 12.7] years) formed the basis of this study. Participants were divided into two groups according to their age (middle-aged, n = 175 and elderly, n = 94). Hypertension and diabetes mellitus were significantly more prevalent and CACS values and HbA1c, NT-proBNP, and GDF-15 levels were significantly higher (all p < 0.001) in the elderly group compared to the middle-aged group. Multivariate ridge regression analysis revealed a significant positive association between GDF-15 and CACS (middle-aged group: β = 0.072, p = 0.333; elderly group: β = 0.148, p = 0.003), and between GDF-15 and ABI (middle-aged group: β = 0.062, p = 0.393; elderly group: β = 0.088, p = 0.041) only in the elderly group. Our results show that GDF-15 is not only a useful biomarker of inflammation but can also predict early signs of asymptomatic atherosclerosis, especially in elderly people with chronic systemic inflammation associated with aging (inflammaging).
Collapse
Affiliation(s)
- Loretta Zsuzsa Kiss
- Heart and Vascular Center, Semmelweis University, 68 Városmajor Street, 1122, Budapest, Hungary.
| | - Balázs Bence Nyárády
- Heart and Vascular Center, Semmelweis University, 68 Városmajor Street, 1122, Budapest, Hungary
| | - Éva Pállinger
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 4 Nagyvárad Square, 1089, Budapest, Hungary
| | - Árpád Lux
- Heart and Vascular Center, Semmelweis University, 68 Városmajor Street, 1122, Budapest, Hungary
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, Netherlands
| | - Ádám Levente Jermendy
- Heart and Vascular Center, Semmelweis University, 68 Városmajor Street, 1122, Budapest, Hungary
| | - Csaba Csobay-Novák
- Heart and Vascular Center, Semmelweis University, 68 Városmajor Street, 1122, Budapest, Hungary
| | - Pál Soós
- Heart and Vascular Center, Semmelweis University, 68 Városmajor Street, 1122, Budapest, Hungary
| | - Zsolt Szelid
- Heart and Vascular Center, Semmelweis University, 68 Városmajor Street, 1122, Budapest, Hungary
| | - Orsolya Láng
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 4 Nagyvárad Square, 1089, Budapest, Hungary
| | - László Kőhidai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, 4 Nagyvárad Square, 1089, Budapest, Hungary
| | - Elek Dinya
- Institute of Digital Health Sciences, Semmelweis University, 15 Ferenc Square, 1094, Budapest, Hungary
| | - Edit Dósa
- Heart and Vascular Center, Semmelweis University, 68 Városmajor Street, 1122, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, 68 Városmajor Street, 1122, Budapest, Hungary
| | - Zsolt Bagyura
- Heart and Vascular Center, Semmelweis University, 68 Városmajor Street, 1122, Budapest, Hungary
| |
Collapse
|
19
|
Adibi JJ, Zhao Y, Koistinen H, Mitchell RT, Barrett ES, Miller R, O'Connor TG, Xun X, Liang HW, Birru R, Smith M, Moog NK. Molecular pathways in placental-fetal development and disruption. Mol Cell Endocrinol 2024; 581:112075. [PMID: 37852527 PMCID: PMC10958409 DOI: 10.1016/j.mce.2023.112075] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/11/2023] [Accepted: 09/24/2023] [Indexed: 10/20/2023]
Abstract
The first trimester of pregnancy ranks high in priority when minimizing harmful exposures, given the wide-ranging types of organogenesis occurring between 4- and 12-weeks' gestation. One way to quantify potential harm to the fetus in the first trimester is to measure a corollary effect on the placenta. Placental biomarkers are widely present in maternal circulation, cord blood, and placental tissue biopsied at birth or at the time of pregnancy termination. Here we evaluate ten diverse pathways involving molecules expressed in the first trimester human placenta based on their relevance to normal fetal development and to the hypothesis of placental-fetal endocrine disruption (perturbation in development that results in abnormal endocrine function in the offspring), namely: human chorionic gonadotropin (hCG), thyroid hormone regulation, peroxisome proliferator activated receptor protein gamma (PPARγ), leptin, transforming growth factor beta, epiregulin, growth differentiation factor 15, small nucleolar RNAs, serotonin, and vitamin D. Some of these are well-established as biomarkers of placental-fetal endocrine disruption, while others are not well studied and were selected based on discovery analyses of the placental transcriptome. A literature search on these biomarkers summarizes evidence of placenta-specific production and regulation of each biomarker, and their role in fetal reproductive tract, brain, and other specific domains of fetal development. In this review, we extend the theory of fetal programming to placental-fetal programming.
Collapse
Affiliation(s)
- Jennifer J Adibi
- Department of Epidemiology, University of Pittsburgh School of Public Health, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Yaqi Zhao
- St. Jude's Research Hospital, Memphis, TN, USA
| | - Hannu Koistinen
- Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Rod T Mitchell
- Department of Paediatric Endocrinology, Royal Hospital for Children and Young People, Edinburgh BioQuarter, Edinburgh, UK
| | - Emily S Barrett
- Environmental and Population Health Bio-Sciences, Rutgers University School of Public Health, Piscataway, NJ, USA
| | - Richard Miller
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas G O'Connor
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
| | - Xiaoshuang Xun
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Hai-Wei Liang
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Rahel Birru
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Megan Smith
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nora K Moog
- Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
20
|
Tarabeih N, Kalinkovich A, Ashkenazi S, Cherny SS, Shalata A, Livshits G. Relationships between Circulating Biomarkers and Body Composition Parameters in Patients with Metabolic Syndrome: A Community-Based Study. Int J Mol Sci 2024; 25:881. [PMID: 38255954 PMCID: PMC10815336 DOI: 10.3390/ijms25020881] [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: 12/06/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Metabolic syndrome (MetS) is a complex disease involving multiple physiological, biochemical, and metabolic abnormalities. The search for reliable biomarkers may help to better elucidate its pathogenesis and develop new preventive and therapeutic strategies. In the present population-based study, we looked for biomarkers of MetS among obesity- and inflammation-related circulating factors and body composition parameters in 1079 individuals (with age range between 18 and 80) belonging to an ethnically homogeneous population. Plasma levels of soluble markers were measured by using ELISA. Body composition parameters were assessed using bioimpedance analysis (BIA). Statistical analysis, including mixed-effects regression, with MetS as a dependent variable, revealed that the most significant independent variables were mainly adipose tissue-related phenotypes, including fat mass/weight (FM/WT) [OR (95% CI)], 2.77 (2.01-3.81); leptin/adiponectin ratio (L/A ratio), 1.50 (1.23-1.83); growth and differentiation factor 15 (GDF-15) levels, 1.32 (1.08-1.62); inflammatory markers, specifically monocyte to high-density lipoprotein cholesterol ratio (MHR), 2.53 (2.00-3.15), and a few others. Additive Bayesian network modeling suggests that age, sex, MHR, and FM/WT are directly associated with MetS and probably affect its manifestation. Additionally, MetS may be causing the GDF-15 and L/A ratio. Our novel findings suggest the existence of complex, age-related, and possibly hierarchical relationships between MetS and factors associated with obesity.
Collapse
Affiliation(s)
- Nader Tarabeih
- Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (N.T.); (S.A.)
| | - Alexander Kalinkovich
- Department of Anatomy and Anthropology, Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (A.K.); (S.S.C.)
| | - Shai Ashkenazi
- Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (N.T.); (S.A.)
| | - Stacey S. Cherny
- Department of Anatomy and Anthropology, Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (A.K.); (S.S.C.)
| | - Adel Shalata
- The Simon Winter Institute for Human Genetics, Bnai Zion Medical Center, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 32000, Israel;
| | - Gregory Livshits
- Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 40700, Israel; (N.T.); (S.A.)
- Department of Anatomy and Anthropology, Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel; (A.K.); (S.S.C.)
| |
Collapse
|
21
|
Fejzo M, Rocha N, Cimino I, Lockhart SM, Petry CJ, Kay RG, Burling K, Barker P, George AL, Yasara N, Premawardhena A, Gong S, Cook E, Rimmington D, Rainbow K, Withers DJ, Cortessis V, Mullin PM, MacGibbon KW, Jin E, Kam A, Campbell A, Polasek O, Tzoneva G, Gribble FM, Yeo GSH, Lam BYH, Saudek V, Hughes IA, Ong KK, Perry JRB, Sutton Cole A, Baumgarten M, Welsh P, Sattar N, Smith GCS, Charnock-Jones DS, Coll AP, Meek CL, Mettananda S, Hayward C, Mancuso N, O'Rahilly S. GDF15 linked to maternal risk of nausea and vomiting during pregnancy. Nature 2024; 625:760-767. [PMID: 38092039 PMCID: PMC10808057 DOI: 10.1038/s41586-023-06921-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 11/30/2023] [Indexed: 01/19/2024]
Abstract
GDF15, a hormone acting on the brainstem, has been implicated in the nausea and vomiting of pregnancy, including its most severe form, hyperemesis gravidarum (HG), but a full mechanistic understanding is lacking1-4. Here we report that fetal production of GDF15 and maternal sensitivity to it both contribute substantially to the risk of HG. We confirmed that higher GDF15 levels in maternal blood are associated with vomiting in pregnancy and HG. Using mass spectrometry to detect a naturally labelled GDF15 variant, we demonstrate that the vast majority of GDF15 in the maternal plasma is derived from the feto-placental unit. By studying carriers of rare and common genetic variants, we found that low levels of GDF15 in the non-pregnant state increase the risk of developing HG. Conversely, women with β-thalassaemia, a condition in which GDF15 levels are chronically high5, report very low levels of nausea and vomiting of pregnancy. In mice, the acute food intake response to a bolus of GDF15 is influenced bi-directionally by prior levels of circulating GDF15 in a manner suggesting that this system is susceptible to desensitization. Our findings support a putative causal role for fetally derived GDF15 in the nausea and vomiting of human pregnancy, with maternal sensitivity, at least partly determined by prepregnancy exposure to the hormone, being a major influence on its severity. They also suggest mechanism-based approaches to the treatment and prevention of HG.
Collapse
Affiliation(s)
- M Fejzo
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - N Rocha
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - I Cimino
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S M Lockhart
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - C J Petry
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - R G Kay
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Peptidomics and Proteomics Core Facility, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - K Burling
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - P Barker
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - A L George
- Peptidomics and Proteomics Core Facility, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - N Yasara
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, Sri Lanka
| | - A Premawardhena
- Adolescent and Adult Thalassaemia Care Center (University Medical Unit), North Colombo Teaching Hospital, Kadawatha, Sri Lanka
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - S Gong
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - E Cook
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - D Rimmington
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - K Rainbow
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - D J Withers
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - V Cortessis
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - P M Mullin
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - K W MacGibbon
- Hyperemesis Education and Research Foundation, Clackamas, OR, USA
| | - E Jin
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - A Kam
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - A Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - O Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - G Tzoneva
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - F M Gribble
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - G S H Yeo
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - B Y H Lam
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - V Saudek
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - I A Hughes
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - K K Ong
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - J R B Perry
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - A Sutton Cole
- Department of Obstetrics and Gynaecology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - M Baumgarten
- Department of Obstetrics and Gynaecology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - P Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - N Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - G C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - D S Charnock-Jones
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - A P Coll
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - C L Meek
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S Mettananda
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, Sri Lanka
- University Paediatrics Unit, Colombo North Teaching Hospital, Ragama, Sri Lanka
| | - C Hayward
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - N Mancuso
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Quantitative and Computational Biology, University of Southern California, California, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, California, CA, USA
| | - S O'Rahilly
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| |
Collapse
|
22
|
Runco DV, DiMeglio LA, Vanderpool CP, Han Y, Daggy J, Kelley MM, Mikesell R, Zimmers TA. Growth differentiation factor 15 (GDF15) elevation in children with newly diagnosed cancer. Front Oncol 2023; 13:1295228. [PMID: 38146512 PMCID: PMC10749306 DOI: 10.3389/fonc.2023.1295228] [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: 09/15/2023] [Accepted: 11/20/2023] [Indexed: 12/27/2023] Open
Abstract
Background Growth differentiation factor 15 (GDF15), an inflammatory marker and mediator of adult cancer cachexia, remains largely unexplored in children. GDF15 increases nausea, vomiting, and anorexia in cancer and contributes to malnutrition, with the potential to be a cachexia therapeutic target. No studies have examined GDF15 in children with newly diagnosed cancer. Our pilot study compares GDF15 in children with newly diagnosed cancer to age- and sex-matched controls and correlates levels with anthropometric measurements and quality of life (QOL). Methods Children with newly diagnosed cancer aged 2-21 years were enrolled with serum GDF15 ELISA, anthropometric measures [height, weight, and mid-upper arm circumference (MUAC)], and QOL assessments (using PedsQL™ Core and Gastrointestinal Modules), which were collected at baseline and repeated 3 months later. Serum GDF15 levels were obtained from age- and sex-matched controls for comparison. Results A total of 57 participants enrolled (N=30, cancer group; N=27, control group) with a median age of 8.8 years (IQR 5.6-15.9 years). The participants were primarily male (54.4%), white (82.5%), and non-Hispanic (82.5%). Cancer diagnoses included acute lymphoblastic leukemia (N=8), lymphoma (N=8), neuroblastoma (N=5), soft tissue tumors (N=4), acute myeloid leukemia (N=2), and single participants with brain, kidney, and bone tumors. Baseline GDF15 was higher in the cancer cohort compared to the control cohort (median=614.6pg/mL and 320.5pg/mL, respectively; p<0.001). When examining participants with evaluable baseline and 3-month follow-up GDF15 levels (N=18), GDF15 was not statistically different (median=657.1pg/mL and 675.3pg/mL, respectively; p=0.702). A total of 13 of the 30 participants and 21 caregivers completed the PedsQL™ Core and Gastrointestinal symptom modules. QOL scores did not differ significantly at 3-month follow-up compared to baseline, but diarrhea worsened (p=0.017). Median participant response for diarrhea at baseline was 92.9 (IQR=92.9-96.4; N=13), which was significantly better than the follow-up (median=78.6; IQR= 71.4-92.9; p=0.017). There were no correlations between change in height, weight, or MUAC and change in GDF15 levels (p=0.351, 0.920, and 0.269 respectively). Conclusion GDF15 was elevated in children with cancer at diagnosis compared to controls but did not correlate with anthropometric measurements or QOL. This pilot study will inform future prospective studies to better describe the natural history of GDF15 and its role in cachexia and as a potential therapeutic target.
Collapse
Affiliation(s)
- Daniel V. Runco
- Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, IN, United States
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Linda A. DiMeglio
- Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, IN, United States
- Division of Pediatric Endocrinology and Diabetology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Charles P. Vanderpool
- Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, IN, United States
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Yan Han
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Joanne Daggy
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mary M. Kelley
- Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, IN, United States
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Raya Mikesell
- Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, IN, United States
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Teresa A. Zimmers
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
23
|
Holmannova D, Borsky P, Parova H, Stverakova T, Vosmik M, Hruska L, Fiala Z, Borska L. Non-Genomic Hallmarks of Aging-The Review. Int J Mol Sci 2023; 24:15468. [PMID: 37895144 PMCID: PMC10607657 DOI: 10.3390/ijms242015468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Aging is a natural, gradual, and inevitable process associated with a series of changes at the molecular, cellular, and tissue levels that can lead to an increased risk of many diseases, including cancer. The most significant changes at the genomic level (DNA damage, telomere shortening, epigenetic changes) and non-genomic changes are referred to as hallmarks of aging. The hallmarks of aging and cancer are intertwined. Many studies have focused on genomic hallmarks, but non-genomic hallmarks are also important and may additionally cause genomic damage and increase the expression of genomic hallmarks. Understanding the non-genomic hallmarks of aging and cancer, and how they are intertwined, may lead to the development of approaches that could influence these hallmarks and thus function not only to slow aging but also to prevent cancer. In this review, we focus on non-genomic changes. We discuss cell senescence, disruption of proteostasis, deregualation of nutrient sensing, dysregulation of immune system function, intercellular communication, mitochondrial dysfunction, stem cell exhaustion and dysbiosis.
Collapse
Affiliation(s)
- Drahomira Holmannova
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (D.H.); (Z.F.); (L.B.)
| | - Pavel Borsky
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (D.H.); (Z.F.); (L.B.)
| | - Helena Parova
- Department of Clinical Biochemistry and Diagnostics, University Hospital, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (H.P.); (T.S.)
| | - Tereza Stverakova
- Department of Clinical Biochemistry and Diagnostics, University Hospital, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (H.P.); (T.S.)
| | - Milan Vosmik
- Department of Oncology and Radiotherapy, University Hospital, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (M.V.); (L.H.)
| | - Libor Hruska
- Department of Oncology and Radiotherapy, University Hospital, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (M.V.); (L.H.)
| | - Zdenek Fiala
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (D.H.); (Z.F.); (L.B.)
| | - Lenka Borska
- Institute of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic; (D.H.); (Z.F.); (L.B.)
| |
Collapse
|
24
|
De Paepe B, Bracke KR, De Bleecker JL. Retrospective Study Shows That Serum Levels of Chemokine CXCL10 and Cytokine GDF15 Support a Diagnosis of Sporadic Inclusion Body Myositis and Immune-Mediated Necrotizing Myopathy. Brain Sci 2023; 13:1369. [PMID: 37891738 PMCID: PMC10605230 DOI: 10.3390/brainsci13101369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
The implementation of novel blood-based biomarkers is desired to reduce the diagnostic delay and burden for myositis patients. In this retrospective study, the potential of C-X-C motif chemokine ligand 10 (CXCL10) and growth differentiation factor 15 (GDF15) was explored in an established patient cohort diagnosed with immune-mediated necrotizing myopathy (IMNM; n = 21), sporadic inclusion body myositis (IBM; n = 18), overlap myositis (OM; n = 3), dermatomyositis (DM; n = 2), and anti-synthetase syndrome (ASS; n = 1), comparing these results with healthy controls (n = 10) and patients with a hereditary neuromuscular disorder (n = 14). CXCL10 and GDF15 were quantified in sera with enzyme-linked immunosorbent assays and immunolocalized in skeletal muscle tissue. In myositis patients, serum CXCL10 levels were significantly increased 9.6-fold compared to healthy controls and 4.2-fold compared to disease controls. Mean levels of CXCL10 were 929 ± 658 pg/mL of serum in IBM and 425 ± 324 pg/mL of serum in IMNM. With the threshold set to 180 pg/mL of CXCL10, myositis patients could be differentiated from healthy and disease controls with a sensitivity of 0.80 and a specificity of 0.71. Incorporating a threshold of 300 pg/mL for GDF15 reduced false negatives to two IMNM patients only. Subsets of muscle-infiltrating immune cells expressed CXCL10, and serum levels correlated with muscle inflammation grade. We propose adding circulating CXCL10 and GDF15 to the blood-based diagnostic toolkit for myositis as a valuable patient-friendly approach.
Collapse
Affiliation(s)
- Boel De Paepe
- Department of Neurology, Ghent University Hospital, B-9000 Ghent, Belgium;
- Neuromuscular Reference Center, Ghent University Hospital, B-9000 Ghent, Belgium
| | - Ken R. Bracke
- Department of Respiratory Medicine, Ghent University Hospital, B-9000 Ghent, Belgium;
| | - Jan L. De Bleecker
- Department of Neurology, Ghent University Hospital, B-9000 Ghent, Belgium;
- Neuromuscular Reference Center, Ghent University Hospital, B-9000 Ghent, Belgium
| |
Collapse
|
25
|
Yaluri N, Stančáková Yaluri A, Žeňuch P, Žeňuchová Z, Tóth Š, Kalanin P. Cardiac Biomarkers and Their Role in Identifying Increased Risk of Cardiovascular Complications in COVID-19 Patients. Diagnostics (Basel) 2023; 13:2508. [PMID: 37568870 PMCID: PMC10417576 DOI: 10.3390/diagnostics13152508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/03/2023] [Accepted: 07/17/2023] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular disease (CVD) is a global health concern, causing significant morbidity and mortality. Both lifestyle and genetics influence the development of CVD. It is often diagnosed late, when the treatment options are limited. Early diagnosis of CVD with help of biomarkers is necessary to prevent adverse outcomes. SARS-CoV-2 infection can cause cardiovascular complications even in patients with no prior history of CVD. This review highlights cardiovascular biomarkers, including novel ones, and their applications as diagnostic and prognostic markers of cardiovascular complications related to SARS-CoV-2 infection. Patients with severe SARS-CoV-2 infection were shown to have elevated levels of cardiac biomarkers, namely N-terminal pro-brain natriuretic peptide (NT-pro-BNP), creatine kinase-myocardial band (CK-MB), and troponins, indicating acute myocardial damage. These biomarkers were also associated with higher mortality rates and therefore should be used throughout COVID-19 patient care to identify high-risk patients promptly to optimize their outcomes. Additionally, microRNAs (miRNAs) are also considered as potential biomarkers and predictors of cardiac and vascular damage in SARS-CoV-2 infection. Identifying molecular pathways contributing to cardiovascular manifestations in COVID-19 is essential for development of early biomarkers, identification of new therapeutic targets, and better prediction and management of cardiovascular outcomes.
Collapse
Affiliation(s)
- Nagendra Yaluri
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | | | - Pavol Žeňuch
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Zuzana Žeňuchová
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Štefan Tóth
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| | - Peter Kalanin
- Center of Clinical and Preclinical Research, University Research Park Medipark, P. J. Šafárik University, 040 01 Košice, Slovakia
| |
Collapse
|
26
|
Fejzo M, Rocha N, Cimino I, Lockhart SM, Petry C, Kay RG, Burling K, Barker P, George AL, Yasara N, Premawardhena A, Gong S, Cook E, Rainbow K, Withers DJ, Cortessis V, Mullin PM, MacGibbon KW, Jin E, Kam A, Campbell A, Polasek O, Tzoneva G, Gribble FM, Yeo G, Lam B, Saudek V, Hughes IA, Ong KK, Perry J, Sutton Cole A, Baumgarten M, Welsh P, Sattar N, Smith G, Charnock Jones DS, Coll AP, Meek CL, Mettananda S, Hayward C, Mancuso N, O'Rahilly S. Fetally-encoded GDF15 and maternal GDF15 sensitivity are major determinants of nausea and vomiting in human pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.542661. [PMID: 37398065 PMCID: PMC10312505 DOI: 10.1101/2023.06.02.542661] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Human pregnancy is frequently accompanied by nausea and vomiting that may become severe and life-threatening, as in hyperemesis gravidarum (HG), the cause of which is unknown. Growth Differentiation Factor-15 (GDF15), a hormone known to act on the hindbrain to cause emesis, is highly expressed in the placenta and its levels in maternal blood rise rapidly in pregnancy. Variants in the maternal GDF15 gene are associated with HG. Here we report that fetal production of GDF15, and maternal sensitivity to it, both contribute substantially to the risk of HG. We found that the great majority of GDF15 in maternal circulation is derived from the feto-placental unit and that higher GDF15 levels in maternal blood are associated with vomiting and are further elevated in patients with HG. Conversely, we found that lower levels of GDF15 in the non-pregnant state predispose women to HG. A rare C211G variant in GDF15 which strongly predisposes mothers to HG, particularly when the fetus is wild-type, was found to markedly impair cellular secretion of GDF15 and associate with low circulating levels of GDF15 in the non-pregnant state. Consistent with this, two common GDF15 haplotypes which predispose to HG were associated with lower circulating levels outside pregnancy. The administration of a long-acting form of GDF15 to wild-type mice markedly reduced subsequent responses to an acute dose, establishing that desensitisation is a feature of this system. GDF15 levels are known to be highly and chronically elevated in patients with beta thalassemia. In women with this disorder, reports of symptoms of nausea or vomiting in pregnancy were strikingly diminished. Our findings support a causal role for fetal derived GDF15 in the nausea and vomiting of human pregnancy, with maternal sensitivity, at least partly determined by pre-pregnancy exposure to GDF15, being a major influence on its severity. They also suggest mechanism-based approaches to the treatment and prevention of HG.
Collapse
Affiliation(s)
- M Fejzo
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - N Rocha
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - I Cimino
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S M Lockhart
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - C Petry
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - R G Kay
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Peptidomics and Proteomics Core Facility, Level 4, Wellcome-MRC Institute of Metabolic Science, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - K Burling
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - P Barker
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Core Biochemical Assay Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - A L George
- Peptidomics and Proteomics Core Facility, Level 4, Wellcome-MRC Institute of Metabolic Science, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - N Yasara
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, 11010, Sri Lanka
| | - A Premawardhena
- Adolescent and Adult Thalassaemia Care Center (University Medical Unit), North Colombo Teaching Hospital, Kadawatha, Sri Lanka
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - S Gong
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - E Cook
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - K Rainbow
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - D J Withers
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - V Cortessis
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
| | - P M Mullin
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - K W MacGibbon
- Hyperemesis Education and Research Foundation, Clackamas, OR
| | - E Jin
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - A Kam
- Department of Obstetrics and Gynaecology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - A Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - O Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - G Tzoneva
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - F M Gribble
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Gsh Yeo
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Byh Lam
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - V Saudek
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - I A Hughes
- Department of Paediatrics, Cambridge University Hospitals NHS Foundation Trust, University of Cambridge, Cambridge, UK
| | - K K Ong
- Department of Paediatrics, Cambridge University Hospitals NHS Foundation Trust, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jrb Perry
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - A Sutton Cole
- Department of Obstetrics and Gynaecology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - M Baumgarten
- Department of Obstetrics and Gynaecology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - P Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - N Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Gcs Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - D S Charnock Jones
- Department of Obstetrics and Gynaecology, University of Cambridge, NIHR Cambridge Biomedical Research Centre, Cambridge, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - A P Coll
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - C L Meek
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - S Mettananda
- Department of Paediatrics, Faculty of Medicine, University of Kelaniya, Thalagolla Road, Ragama, 11010, Sri Lanka
- University Paediatrics Unit, Colombo North Teaching Hospital, Ragama, Sri Lanka
| | - C Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU,16, UK
| | - N Mancuso
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
- Department of Quantitative and Computational Biology, University of Southern California
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California
| | - S O'Rahilly
- Medical Research Council (MRC) Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Al-Sawaf O, Weiss J, Skrzypski M, Lam JM, Karasaki T, Zambrana F, Kidd AC, Frankell AM, Watkins TBK, Martínez-Ruiz C, Puttick C, Black JRM, Huebner A, Bakir MA, Sokač M, Collins S, Veeriah S, Magno N, Naceur-Lombardelli C, Prymas P, Toncheva A, Ward S, Jayanth N, Salgado R, Bridge CP, Christiani DC, Mak RH, Bay C, Rosenthal M, Sattar N, Welsh P, Liu Y, Perrimon N, Popuri K, Beg MF, McGranahan N, Hackshaw A, Breen DM, O'Rahilly S, Birkbak NJ, Aerts HJWL, Jamal-Hanjani M, Swanton C. Body composition and lung cancer-associated cachexia in TRACERx. Nat Med 2023; 29:846-858. [PMID: 37045997 PMCID: PMC7614477 DOI: 10.1038/s41591-023-02232-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 01/24/2023] [Indexed: 04/14/2023]
Abstract
Cancer-associated cachexia (CAC) is a major contributor to morbidity and mortality in individuals with non-small cell lung cancer. Key features of CAC include alterations in body composition and body weight. Here, we explore the association between body composition and body weight with survival and delineate potential biological processes and mediators that contribute to the development of CAC. Computed tomography-based body composition analysis of 651 individuals in the TRACERx (TRAcking non-small cell lung Cancer Evolution through therapy (Rx)) study suggested that individuals in the bottom 20th percentile of the distribution of skeletal muscle or adipose tissue area at the time of lung cancer diagnosis, had significantly shorter lung cancer-specific survival and overall survival. This finding was validated in 420 individuals in the independent Boston Lung Cancer Study. Individuals classified as having developed CAC according to one or more features at relapse encompassing loss of adipose or muscle tissue, or body mass index-adjusted weight loss were found to have distinct tumor genomic and transcriptomic profiles compared with individuals who did not develop such features. Primary non-small cell lung cancers from individuals who developed CAC were characterized by enrichment of inflammatory signaling and epithelial-mesenchymal transitional pathways, and differentially expressed genes upregulated in these tumors included cancer-testis antigen MAGEA6 and matrix metalloproteinases, such as ADAMTS3. In an exploratory proteomic analysis of circulating putative mediators of cachexia performed in a subset of 110 individuals from TRACERx, a significant association between circulating GDF15 and loss of body weight, skeletal muscle and adipose tissue was identified at relapse, supporting the potential therapeutic relevance of targeting GDF15 in the management of CAC.
Collapse
Affiliation(s)
- Othman Al-Sawaf
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Jakob Weiss
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Diagnostic and Interventional Radiology, University Freiburg, Freiburg, Germany
| | - Marcin Skrzypski
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Jie Min Lam
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Oncology, University College London Hospitals, London, UK
| | - Takahiro Karasaki
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | | | - Andrew C Kidd
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Thomas B K Watkins
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Carlos Martínez-Ruiz
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Clare Puttick
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - James R M Black
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Ariana Huebner
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Maise Al Bakir
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Mateo Sokač
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Susie Collins
- Early Clinical Development, Pfizer UK Ltd, Cambridge, UK
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Neil Magno
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | - Paulina Prymas
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Antonia Toncheva
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Sophia Ward
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Nick Jayanth
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Roberto Salgado
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - David C Christiani
- Department of Medicine, Massachusetts General Hospital/Harvard Medicine School, and Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Raymond H Mak
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Camden Bay
- Department of Radiology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA, USA
| | - Michael Rosenthal
- Department of Radiology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA, USA
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Paul Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Ying Liu
- Department of Genetics, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Burnaby, Canada
| | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, British Colombia, Canada
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Allan Hackshaw
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Danna M Breen
- Internal Medicine Research Unit, Pfizer, Cambridge, MA, USA
| | - Stephen O'Rahilly
- Wellcome Trust-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Nicolai J Birkbak
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Hugo J W L Aerts
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, Maastricht, The Netherlands
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
28
|
deFilippi CR, Tran H, Gattani R, Daniels LB, Shah P, Ilkhanoff L, Christenson R, Lima JA, Seliger S. Association of cardiac troponin T and growth differentiation factor 15 with replacement and interstitial cardiac fibrosis in community dwelling adults: The multi-ethnic study of atherosclerosis. Front Cardiovasc Med 2023; 10:1104715. [PMID: 36844723 PMCID: PMC9949377 DOI: 10.3389/fcvm.2023.1104715] [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: 12/07/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Background Subclinical abnormalities in myocardial structure (stage B heart failure) may be identified by cardiac and non-organ specific biomarkers. The associations of high-sensitivity cardiac troponin T (hs-cTnT) and growth differentiation factor-15 (GDF-15) with cardiac magnetic resonance imaging (CMR) interstitial fibrosis (extracellular volume [ECV]) is unknown and for GDF-15 the association with replacement (late gadolinium enhancement [LGE]) is also unknown. GDF-15 is a systemic biomarker also released by myocytes associated with fibrosis and inflammation. We sought to define the associations of hs-cTnT and GDF-15 with these CMR fibrosis measures in the MESA cohort. Methods We measured hs-cTnT and GDF-15 in MESA participants free of cardiovascular disease at exam 5. CMR measurements were complete in 1737 for LGE and 1258 for ECV assessment. We estimated the association of each biomarker with LGE and increased ECV (4th quartile) using logistic regression, adjusted for demographics and risk factors. Results Mean age of the participants was 68 ± 9 years. Unadjusted, both biomarkers were associated with LGE, but after adjustment only hs-cTnT concentrations remained significant (4th vs. 1st quartile OR] 7.5, 95% CI: 2.1, 26.6). For interstitial fibrosis both biomarkers were associated with 4th quartile ECV, but the association was attenuated compared to replacement fibrosis. After adjustment, only hs-cTnT concentrations remained significant (1st to 4th quartile OR 1.7, 95%CI: 1.1, 2.8). Conclusion Our findings identify that both interstitial and replacement fibrosis are associated with myocyte cell death/injury, but GDF-15 a non-organ specific biomarker prognostic for incident cardiovascular disease is not associated with preclinical evidence of cardiac fibrosis.
Collapse
Affiliation(s)
- Christopher R. deFilippi
- Inova Heart and Vascular Institute, Falls Church, VA, United States,*Correspondence: Christopher R. deFilippi,
| | - Henry Tran
- Inova Heart and Vascular Institute, Falls Church, VA, United States
| | - Raghav Gattani
- Inova Heart and Vascular Institute, Falls Church, VA, United States
| | - Lori B. Daniels
- Division of Cardiology, University of California and San Diego Medical Center, San Diego, CA, United States
| | - Palak Shah
- Inova Heart and Vascular Institute, Falls Church, VA, United States
| | | | - Robert Christenson
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Joao A. Lima
- The Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Stephen Seliger
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|