1
|
Tepetes NI, Kourek C, Papamichail A, Xanthopoulos A, Kostakou P, Paraskevaidis I, Briasoulis A. Transition to Advanced Heart Failure: From Identification to Improving Prognosis. J Cardiovasc Dev Dis 2025; 12:104. [PMID: 40137102 PMCID: PMC11943400 DOI: 10.3390/jcdd12030104] [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: 02/15/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Advanced heart failure (AHF) represents the terminal stage of heart failure (HF), characterized by persistent symptoms and functional limitations despite optimal guideline-directed medical therapy (GDMT). This review explores the clinical definition, pathophysiology, and therapeutic approaches for AHF. Characterized by severe symptoms, New York Heart Association (NYHA) class III-IV, significant cardiac dysfunction, and frequent hospitalizations, AHF presents substantial challenges in prognosis and management. Pathophysiological mechanisms include neurohormonal activation, ventricular remodeling, and systemic inflammation, leading to reduced cardiac output and organ dysfunction. Therapeutic strategies for AHF involve a multidisciplinary approach, including pharmacological treatments, device-based interventions like ventricular assisted devices, and advanced options such as heart transplantation. Despite progress, AHF management faces limitations, including disparities in access to care and the need for personalized approaches. Novel therapies, artificial intelligence, and remote monitoring technologies offer future opportunities to improve outcomes. Palliative care, which focuses on symptom relief and quality of life, remains crucial for patients ineligible for invasive interventions. Early identification and timely intervention are pivotal for enhancing survival and functional outcomes in this vulnerable population. This review underscores the necessity of integrating innovative technologies, personalized medicine, and robust palliative strategies into AHF management to address its high morbidity and mortality.
Collapse
Affiliation(s)
- Nikolaos-Iason Tepetes
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (N.-I.T.); (P.K.)
| | - Christos Kourek
- Department of Cardiology, 417 Army Share Fund Hospital of Athens (NIMTS), 11521 Athens, Greece;
| | - Adamantia Papamichail
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Andrew Xanthopoulos
- Department of Cardiology, University Hospital of Larissa, 41110 Larissa, Greece;
| | - Peggy Kostakou
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (N.-I.T.); (P.K.)
| | | | - Alexandros Briasoulis
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (N.-I.T.); (P.K.)
| |
Collapse
|
2
|
Bastos JM, Colaço B, Baptista R, Gavina C, Vitorino R. Innovations in heart failure management: The role of cutting-edge biomarkers and multi-omics integration. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100290. [PMID: 40129519 PMCID: PMC11930597 DOI: 10.1016/j.jmccpl.2025.100290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/11/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
Heart failure (HF) remains a major cause of morbidity and mortality worldwide and represents a major challenge for diagnosis, prognosis and treatment due to its heterogeneity. Traditional biomarkers such as BNP and NT-proBNP are valuable but insufficient to capture the complexity of HF, especially phenotypes such as HF with preserved ejection fraction (HFpEF). Recent advances in multi-omics technology and novel biomarkers such as cell-free DNA (cfDNA), microRNAs (miRNAs), ST2 and galectin-3 offer transformative potential for HF management. This review explores the integration of these innovative biomarkers into clinical practice and highlights their benefits, such as improved diagnostic accuracy, enhanced risk stratification and non-invasive monitoring capabilities. By leveraging multi-omics approaches, including lipidomics and metabolomics, clinicians can uncover new pathways, refine the classification of HF phenotypes, and develop personalized therapeutic strategies tailored to individual patient profiles. Remarkable advances in proteomics and metabolomics have identified biomarkers associated with key HF mechanisms such as mitochondrial dysfunction, inflammation and fibrosis, paving the way for targeted therapies and early interventions. Despite the promising results, significant challenges remain in translating these findings into routine care, including high costs, technical limitations and the need for large-scale validation studies. This report argues for an integrative, multi-omics-based model to overcome these obstacles and emphasizes the importance of collaboration between researchers, clinicians and policy makers. By linking innovative science with practical applications, multi-omics approaches have the potential to redefine HF management and lead to better patient outcomes and more sustainable healthcare systems.
Collapse
Affiliation(s)
- Jose Mesquita Bastos
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
- Cardiology Department, Hospital Infante D. Pedro, Centro Hospitalar do Baixo Vouga, Aveiro, Portugal
| | - Beatriz Colaço
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
- Cardiology Department, Hospital Infante D. Pedro, Centro Hospitalar do Baixo Vouga, Aveiro, Portugal
| | - Rui Baptista
- Department of Cardiology, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Cristina Gavina
- Pedro Hispano Hospital - ULS Matosinhos, Matosinhos, Portugal
- Cardiology Department, Faculty of Medicine, University of Porto, Oporto, Portugal
- RISE- Health Research Network, Faculty of Medicine, University of Porto, Oporto, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
3
|
Ge X, Brittain B, Dawson L, Dwivedi G, Kaye DM, Morahan G. A Genetic Test to Identify People at High Risk of Heart Failure. Int J Mol Sci 2025; 26:1782. [PMID: 40004245 PMCID: PMC11855781 DOI: 10.3390/ijms26041782] [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: 12/19/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Earlier intervention may delay or prevent heart failure (HF), a widespread health problem. However, it is not currently possible to identify those who are most at risk, especially before the appearance of any clinical signs. This study presents the development and subsequent validation of a novel genetic test for predicting the risk of HF, utilizing data from three independent cohorts of Australian and US subjects. We developed a first-phase test using the Baker Biobank case-control cohort, identifying 41 genetic variants indicative of HF risk through genome-wide interaction and association analyses. Subsequently, a second-phase test was designed. This identified 29 additional single-nucleotide polymorphisms. The combination of these two tests resulted in an aggregate test with a high predictive accuracy, achieving an Area Under the Curve of 0.93 and a balanced accuracy of 0.89. High genetic risk subjects in the Baker Biobank cohort had an odds ratio of 533.2. The test's robustness was validated by applying it to data from the Busselton Health Study and the Atherosclerosis Risk in Communities cohorts, yielding, respectively, Areas Under the Curve of 0.83 and 0.72, a balanced accuracy of 0.76 and 0.67, and Odds Ratios of 12.3 and 4.6. These results highlight the critical role of genetic factors in the development of heart failure and demonstrate this test's potential as a significant tool for clinical HF risk prediction.
Collapse
Affiliation(s)
- Xintian Ge
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia; (X.G.); (B.B.); (G.D.)
- Stroke Research Centre, Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Bek Brittain
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia; (X.G.); (B.B.); (G.D.)
| | - Luke Dawson
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (L.D.); (D.M.K.)
| | - Girish Dwivedi
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia; (X.G.); (B.B.); (G.D.)
- Department of Cardiology, Fiona Stanley Hospital, Perth, WA 6150, Australia
| | - David M. Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia; (L.D.); (D.M.K.)
| | - Grant Morahan
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, Nedlands, WA 6009, Australia; (X.G.); (B.B.); (G.D.)
- Advanced Genetic Diagnostics, Nedlands, WA 6009, Australia
| |
Collapse
|
4
|
Ferreira M, Geraldes V, Felix AC, Oliveira M, Laranjo S, Rocha I. Advancing Atrial Fibrillation Research: The Role of Animal Models, Emerging Technologies and Translational Challenges. Biomedicines 2025; 13:307. [PMID: 40002720 PMCID: PMC11853233 DOI: 10.3390/biomedicines13020307] [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: 12/20/2024] [Revised: 01/21/2025] [Accepted: 01/25/2025] [Indexed: 02/27/2025] Open
Abstract
Atrial fibrillation (AF) is the most prevalent sustained cardiac arrhythmia, presenting a significant global healthcare challenge due to its rising incidence, association with increased morbidity and mortality, and economic burden. This arrhythmia is driven by a complex interplay of electrical, structural, and autonomic remodelling, compounded by genetic predisposition, systemic inflammation, and oxidative stress. Despite advances in understanding its pathophysiology, AF management remains suboptimal, with ongoing debates surrounding rhythm control, rate control, and anticoagulation strategies. Animal models have been instrumental in elucidating AF mechanisms, facilitating preclinical research, and advancing therapeutic development. This review critically evaluates the role of animal models in studying AF, emphasizing their utility in exploring electrical, structural, and autonomic remodelling. It highlights the strengths and limitations of various models, from rodents to large animals, in replicating human AF pathophysiology and advancing translational research. Emerging approaches, including optogenetics, advanced imaging, computational modelling, and tissue engineering, are reshaping AF research, bridging the gap between preclinical and clinical applications. We also briefly discuss ethical considerations, the translational challenges of animal studies and future directions, including integrative multi-species approaches, omics technologies and personalized computational models. By addressing these challenges and addressing emerging methodologies, this review underscores the importance of refining experimental models and integrating innovative technologies to improve AF management and outcomes.
Collapse
Affiliation(s)
- Monica Ferreira
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal; (M.F.); (V.G.); (M.O.)
- Centro Cardiovascular da Universidade de Lisboa-CCUL, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Vera Geraldes
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal; (M.F.); (V.G.); (M.O.)
- Centro Cardiovascular da Universidade de Lisboa-CCUL, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Ana Clara Felix
- Pediatric Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal; (A.C.F.); (S.L.)
| | - Mario Oliveira
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal; (M.F.); (V.G.); (M.O.)
- Centro Cardiovascular da Universidade de Lisboa-CCUL, Universidade de Lisboa, 1649-004 Lisbon, Portugal
- Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal
| | - Sergio Laranjo
- Pediatric Cardiology Department, Hospital de Santa Marta, Unidade Local de Saúde de S. José, 1150-199 Lisbon, Portugal; (A.C.F.); (S.L.)
- CHRC, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal
| | - Isabel Rocha
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal; (M.F.); (V.G.); (M.O.)
- Centro Cardiovascular da Universidade de Lisboa-CCUL, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| |
Collapse
|
5
|
Ahmed Z. Applying AI/ML for Analyzing Gene Expression Patterns. Methods Mol Biol 2025; 2880:319-330. [PMID: 39900767 DOI: 10.1007/978-1-0716-4276-4_16] [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] [Indexed: 02/05/2025]
Abstract
Artificial intelligence (AI) and machine learning (ML) have advanced in several areas and fields of life; however, its progress in the field of genomics is not matching the levels others have achieved. Challenges include but are not limited to the handling and analysis of high volumes of complex genomic data, and the expertise needed to implement and execute AI/ML approaches. In this chapter, we highlight the importance of transcriptomics, and RNA-seq driven gene expression data exploration to discover novel biomarkers and predict rare, common, and complex diseases. We discuss relevant high volume sequence data generated in the recent past and its availability through various channels, development of orthodox bioinformatics tools and technologies to investigate significantly expressed and abundantly enriched genes, and the implementation of cutting-edge AI/ML approaches to observe disease specific patterns. Current challenges include but are not limited to the acceptance of AI/ML in the scientific research and clinical environments, especially in providing personalized diagnoses and treatments. Reasons include unavailability of user-friendly AI/ML applications and reproducible results. Addressing these issues, we discuss our recently developed Findable, Accessible, Intelligent, and Reproducible (FAIR) solutions, designed for the users with and without computational background to discover biomarkers and predict diseases with high accuracy. We strongly believe that the rightful application of AI/ML techniques has the potential to open avenues for broader research, ultimately leading to personalized interventions and novel treatment targets. Its widespread application will contribute to the public health at large in the United States and around the globe.
Collapse
Affiliation(s)
- Zeeshan Ahmed
- Department of Medicine, Division of Cardiovascular Disease and Hypertension, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers Health, New Brunswick, NJ, USA.
| |
Collapse
|
6
|
Narayanan R, DeGroat W, Peker E, Zeeshan S, Ahmed Z. VAREANT: a bioinformatics application for gene variant reduction and annotation. BIOINFORMATICS ADVANCES 2024; 5:vbae210. [PMID: 39927292 PMCID: PMC11802749 DOI: 10.1093/bioadv/vbae210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/08/2024] [Accepted: 12/30/2024] [Indexed: 02/11/2025]
Abstract
Motivation The analysis of high-quality genomic variant data may offer a more complete understanding of the human genome, enabling researchers to identify novel biomarkers, stratify patients based on disease risk factors, and decipher underlying biological pathways. Although the availability of genomic data has sharply increased in recent years, the accessibility of bioinformatic tools to aid in its preparation is still lacking. Limitations with processing genomic data primarily include its large volume, associated computational and storage costs, and difficulty in identifying targeted and relevant information. Results We present VAREANT, an accessible and configurable bioinformatic application to support the preparation of variant data into a usable analysis-ready format. VAREANT is comprised of three standalone modules: (i) Pre-processing, (ii) Variant Annotation, (iii) AI/ML Data Preparation. Pre-processing supports the fine-grained filtering of complex variant datasets to eliminate extraneous data. Variant Annotation allows for the addition of variant metadata from the latest public annotation databases for subsequent analysis and interpretation. AI/ML Data Preparation supports the user in creating AI/ML-ready datasets suitable for immediate analysis with minimal pre-processing required. We have successfully tested and validated our tool on numerous variable-sized datasets and implemented VAREANT in two case studies involving patients with cardiovascular diseases. Availability and implementation The open-source code of VAREANT is available at GitHub: https://github.com/drzeeshanahmed/Gene_VAREANT.
Collapse
Affiliation(s)
- Rishabh Narayanan
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States
| | - William DeGroat
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States
| | - Elizabeth Peker
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States
| | - Saman Zeeshan
- Department of Biomedical and Health Informatics, UMKC School of Medicine, Kansas City, MO 64108, United States
| | - Zeeshan Ahmed
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States
- Department of Medicine, Division of Cardiovascular Diseases and Hypertension, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, United States
| |
Collapse
|
7
|
Wang Y, Chatterjee E, Li G, Xu J, Xiao J. Force-sensing protein expression in response to cardiovascular mechanotransduction. EBioMedicine 2024; 110:105412. [PMID: 39481337 PMCID: PMC11554632 DOI: 10.1016/j.ebiom.2024.105412] [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: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Force-sensing biophysical cues in microenvironment, including extracellular matrix performances, stretch-mediated mechanics, shear stress and flow-induced hemodynamics, have a significant influence in regulating vascular morphogenesis and cardiac remodeling by mechanotransduction. Once cells perceive these extracellular mechanical stimuli, Piezo activation promotes calcium influx by forming integrin-adhesion-coupling receptors. This induces robust contractility of cytoskeleton structures to further transmit biomechanical alternations into nuclei by regulating Hippo-Yes associated protein (YAP) signaling pathway between cytoplasmic and nuclear translocation. Although biomechanical stimuli are widely studied in cardiovascular diseases, the expression of force-sensing proteins in response to cardiovascular mechanotransduction has not been systematically concluded. Therefore, this review will summarize the force-sensing Piezo, cytoskeleton and YAP proteins to mediate extracellular mechanics, and also give the prominent emphasis on intrinsic connection of these mechanical proteins and cardiovascular mechanotransduction. Extensive insights into cardiovascular mechanics may provide some new strategies for cardiovascular clinical therapy.
Collapse
Affiliation(s)
- Yongtao Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jiahong Xu
- Department of Cardiology, Shanghai Gongli Hospital, Shanghai 200135, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
8
|
DeGroat W, Abdelhalim H, Peker E, Sheth N, Narayanan R, Zeeshan S, Liang BT, Ahmed Z. Multimodal AI/ML for discovering novel biomarkers and predicting disease using multi-omics profiles of patients with cardiovascular diseases. Sci Rep 2024; 14:26503. [PMID: 39489837 PMCID: PMC11532369 DOI: 10.1038/s41598-024-78553-6] [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: 06/07/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024] Open
Abstract
Cardiovascular diseases (CVDs) are complex, multifactorial conditions that require personalized assessment and treatment. Advancements in multi-omics technologies, namely RNA sequencing and whole-genome sequencing, have provided translational researchers with a comprehensive view of the human genome. The efficient synthesis and analysis of this data through integrated approach that characterizes genetic variants alongside expression patterns linked to emerging phenotypes, can reveal novel biomarkers and enable the segmentation of patient populations based on personalized risk factors. In this study, we present a cutting-edge methodology rooted in the integration of traditional bioinformatics, classical statistics, and multimodal machine learning techniques. Our approach has the potential to uncover the intricate mechanisms underlying CVD, enabling patient-specific risk and response profiling. We sourced transcriptomic expression data and single nucleotide polymorphisms (SNPs) from both CVD patients and healthy controls. By integrating these multi-omics datasets with clinical demographic information, we generated patient-specific profiles. Utilizing a robust feature selection approach, we identified a signature of 27 transcriptomic features and SNPs that are effective predictors of CVD. Differential expression analysis, combined with minimum redundancy maximum relevance feature selection, highlighted biomarkers that explain the disease phenotype. This approach prioritizes both biological relevance and efficiency in machine learning. We employed Combination Annotation Dependent Depletion scores and allele frequencies to identify variants with pathogenic characteristics in CVD patients. Classification models trained on this signature demonstrated high-accuracy predictions for CVD. The best performing of these models was an XGBoost classifier optimized via Bayesian hyperparameter tuning, which was able to correctly classify all patients in our test dataset. Using SHapley Additive exPlanations, we created risk assessments for patients, offering further contextualization of these predictions in a clinical setting. Across the cohort, RPL36AP37 and HBA1 were scored as the most important biomarkers for predicting CVDs. A comprehensive literature review revealed that a substantial portion of the diagnostic biomarkers identified have previously been associated with CVD. The framework we propose in this study is unbiased and generalizable to other diseases and disorders.
Collapse
Affiliation(s)
- William DeGroat
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers, The State University of New Jersey, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Habiba Abdelhalim
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers, The State University of New Jersey, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Elizabeth Peker
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers, The State University of New Jersey, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Neev Sheth
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers, The State University of New Jersey, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Rishabh Narayanan
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers, The State University of New Jersey, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Saman Zeeshan
- Department of Biomedical and Health Informatics, UMKC School of Medicine, 2411 Holmes Street, Kansas City, MO, 64108, USA
| | - Bruce T Liang
- Pat and Jim Calhoun Cardiology Center, UConn Health, 263 Farmington Ave, Farmington, CT, USA
- UConn School of Medicine, University of Connecticut, 263 Farmington Ave, Farmington, CT, USA
| | - Zeeshan Ahmed
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers, The State University of New Jersey, 112 Paterson St, New Brunswick, NJ, 08901, USA.
- UConn School of Medicine, University of Connecticut, 263 Farmington Ave, Farmington, CT, USA.
- Department of Medicine, Division of Cardiovascular Disease and Hypertension, Robert Wood Johnson Medical School, Rutgers Health, 125 Paterson St, New Brunswick, NJ, 08901, USA.
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
9
|
Duenas S, McGee Z, Mhatre I, Mayilvahanan K, Patel KK, Abdelhalim H, Jayprakash A, Wasif U, Nwankwo O, Degroat W, Yanamala N, Sengupta PP, Fine D, Ahmed Z. Computational approaches to investigate the relationship between periodontitis and cardiovascular diseases for precision medicine. Hum Genomics 2024; 18:116. [PMID: 39427205 PMCID: PMC11491019 DOI: 10.1186/s40246-024-00685-7] [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: 04/06/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
Periodontitis is a highly prevalent inflammatory illness that leads to the destruction of tooth supporting tissue structures and has been associated with an increased risk of cardiovascular disease (CVD). Precision medicine, an emerging branch of medical treatment, aims can further improve current traditional treatment by personalizing care based on one's environment, genetic makeup, and lifestyle. Genomic databases have paved the way for precision medicine by elucidating the pathophysiology of complex, heritable diseases. Therefore, the investigation of novel periodontitis-linked genes associated with CVD will enhance our understanding of their linkage and related biochemical pathways for targeted therapies. In this article, we highlight possible mechanisms of actions connecting PD and CVD. Furthermore, we delve deeper into certain heritable inflammatory-associated pathways linking the two. The goal is to gather, compare, and assess high-quality scientific literature alongside genomic datasets that seek to establish a link between periodontitis and CVD. The scope is focused on the most up to date and authentic literature published within the last 10 years, indexed and available from PubMed Central, that analyzes periodontitis-associated genes linked to CVD. Based on the comparative analysis criteria, fifty-one genes associated with both periodontitis and CVD were identified and reported. The prevalence of genes associated with both CVD and periodontitis warrants investigation to assess the validity of a potential linkage between the pathophysiology of both diseases.
Collapse
Affiliation(s)
- Sophia Duenas
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Zachary McGee
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Ishani Mhatre
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Karthikeyan Mayilvahanan
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Kush Ketan Patel
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Habiba Abdelhalim
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Atharv Jayprakash
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Uzayr Wasif
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Oluchi Nwankwo
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - William Degroat
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Naveena Yanamala
- Division of Cardiovascular Diseases and Hypertension, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA
- Department of Medicine, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA
| | - Partho P Sengupta
- Division of Cardiovascular Diseases and Hypertension, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA
- Department of Medicine, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA
| | - Daniel Fine
- Department of Oral Biology, Rutgers School of Dental Medicine, 110 Bergen Street, Newark, NJ, US
| | - Zeeshan Ahmed
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA.
- Division of Cardiovascular Diseases and Hypertension, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA.
- Department of Medicine, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, 125 Paterson St, New Brunswick, NJ, USA.
| |
Collapse
|
10
|
Wu J, Ouyang P, Huang R, Cui Y, Yang Z, Xu W, Ma R, Xiang G, Zeng W, Wu W, Li J. METTL16 Promotes Stability of SYNPO2L mRNA and leading to Cancer Cell Lung Metastasis by Secretion of COL10A1 and attract the Cancer-Associated Fibroblasts. Int J Biol Sci 2024; 20:4128-4145. [PMID: 39247832 PMCID: PMC11379079 DOI: 10.7150/ijbs.95375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/11/2024] [Indexed: 09/10/2024] Open
Abstract
The occurrence of metastasis is a major factor contributing to poor prognosis in colorectal cancer. Different stages of the disease play a crucial role in distant metastasis. Furthermore, m6A has been demonstrated to play a significant role in regulating tumor metastasis. Therefore, we conducted an analysis of transcriptome data from high-stage and low-stage colorectal cancer patients in The Cancer Genome Atlas (TCGA) to identify genes associated with m6A-related regulation. We identified SYNPO2L as a core gene regulated by m6A, and it is correlated with adverse prognosis and metastasis in patients. Additionally, we demonstrated that the m6A writer gene Mettl16 can regulate the stability of SYNPO2L through interaction with YTHDC1. Subsequently, using Weighted Gene Co-expression Network Analysis (WGCNA), we discovered that SYNPO2L can regulate COL10A1, mediating the actions of Cancer-Associated Fibroblasts. SYNPO2L promotes the secretion of COL10A1 and the infiltration of tumor-associated fibroblasts, thereby facilitating Epithelial-Mesenchymal Transition (EMT) in tumor cells and making them more prone to distant metastasis.
Collapse
Affiliation(s)
- Jianlong Wu
- Department of General Surgery, Zhongshan City People's Hospital, Zhongshan, Guangdong 528403, P.R. China
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Peng Ouyang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Rui Huang
- Department of Gastrointestinal surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong 510317, P.R. China
| | - Yao Cui
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R. China
| | - Zhihao Yang
- Department of Biological Science, Mellon College of Science, Carnegie Mellon University. 5000 Forbes Avenue, Pittsburgh, PA, USA 15213
| | - Wei Xu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Rui Ma
- College of Bioengineering, Chongqing University, Chongqing 400044, P.R. China
| | - Guoan Xiang
- Department of Gastrointestinal surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong 510317, P.R. China
| | - Wei Zeng
- Department of General Surgery, Yiyang Central Hospital Affiliated to Hunan University of Chinese Medicine, Yiyang, Hunan 413000, P.R. China
| | - Wang Wu
- College of Bioengineering, Chongqing University, Chongqing 400044, P.R. China
- Institute of Burn Research, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jian Li
- Department of General Surgery, Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
- Department of General Surgery, Henan Cancer Hospital, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
11
|
Mitrokhin V, Hadzi-Petrushev N, Kazanski V, Schileyko S, Kamkina O, Rodina A, Zolotareva A, Zolotarev V, Kamkin A, Mladenov M. The Role of K ACh Channels in Atrial Fibrillation. Cells 2024; 13:1014. [PMID: 38920645 PMCID: PMC11201540 DOI: 10.3390/cells13121014] [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: 05/16/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
This manuscript explores the intricate role of acetylcholine-activated inward rectifier potassium (KACh) channels in the pathogenesis of atrial fibrillation (AF), a common cardiac arrhythmia. It delves into the molecular and cellular mechanisms that underpin AF, emphasizing the vital function of KACh channels in modulating the atrial action potential and facilitating arrhythmogenic conditions. This study underscores the dual nature of KACh activation and its genetic regulation, revealing that specific variations in potassium channel genes, such as Kir3.4 and K2P3.1, significantly influence the electrophysiological remodeling associated with AF. Furthermore, this manuscript identifies the crucial role of the KACh-mediated current, IKACh, in sustaining arrhythmia through facilitating shorter re-entry circuits and stabilizing the re-entrant circuits, particularly in response to vagal nerve stimulation. Experimental findings from animal models, which could not induce AF in the absence of muscarinic activation, highlight the dependency of AF induction on KACh channel activity. This is complemented by discussions on therapeutic interventions, where KACh channel blockers have shown promise in AF management. Additionally, this study discusses the broader implications of KACh channel behavior, including its ubiquitous presence across different cardiac regions and species, contributing to a comprehensive understanding of AF dynamics. The implications of these findings are profound, suggesting that targeting KACh channels might offer new therapeutic avenues for AF treatment, particularly in cases resistant to conventional approaches. By integrating genetic, cellular, and pharmacological perspectives, this manuscript offers a holistic view of the potential mechanisms and therapeutic targets in AF, making a significant contribution to the field of cardiac arrhythmia research.
Collapse
Affiliation(s)
- Vadim Mitrokhin
- Institute of Physiology, Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov, Russian National Research Medical University” Ministry of Health, 117997 Moscow, Russia; (V.M.); (V.K.); (S.S.); (O.K.); (A.R.); (A.Z.); (V.Z.); (A.K.)
| | - Nikola Hadzi-Petrushev
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia;
| | - Viktor Kazanski
- Institute of Physiology, Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov, Russian National Research Medical University” Ministry of Health, 117997 Moscow, Russia; (V.M.); (V.K.); (S.S.); (O.K.); (A.R.); (A.Z.); (V.Z.); (A.K.)
| | - Stanislav Schileyko
- Institute of Physiology, Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov, Russian National Research Medical University” Ministry of Health, 117997 Moscow, Russia; (V.M.); (V.K.); (S.S.); (O.K.); (A.R.); (A.Z.); (V.Z.); (A.K.)
| | - Olga Kamkina
- Institute of Physiology, Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov, Russian National Research Medical University” Ministry of Health, 117997 Moscow, Russia; (V.M.); (V.K.); (S.S.); (O.K.); (A.R.); (A.Z.); (V.Z.); (A.K.)
| | - Anastasija Rodina
- Institute of Physiology, Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov, Russian National Research Medical University” Ministry of Health, 117997 Moscow, Russia; (V.M.); (V.K.); (S.S.); (O.K.); (A.R.); (A.Z.); (V.Z.); (A.K.)
| | - Alexandra Zolotareva
- Institute of Physiology, Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov, Russian National Research Medical University” Ministry of Health, 117997 Moscow, Russia; (V.M.); (V.K.); (S.S.); (O.K.); (A.R.); (A.Z.); (V.Z.); (A.K.)
| | - Valentin Zolotarev
- Institute of Physiology, Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov, Russian National Research Medical University” Ministry of Health, 117997 Moscow, Russia; (V.M.); (V.K.); (S.S.); (O.K.); (A.R.); (A.Z.); (V.Z.); (A.K.)
| | - Andre Kamkin
- Institute of Physiology, Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov, Russian National Research Medical University” Ministry of Health, 117997 Moscow, Russia; (V.M.); (V.K.); (S.S.); (O.K.); (A.R.); (A.Z.); (V.Z.); (A.K.)
| | - Mitko Mladenov
- Institute of Physiology, Federal State Autonomous Educational Institution of Higher Education “N.I. Pirogov, Russian National Research Medical University” Ministry of Health, 117997 Moscow, Russia; (V.M.); (V.K.); (S.S.); (O.K.); (A.R.); (A.Z.); (V.Z.); (A.K.)
- Institute of Biology, Faculty of Natural Sciences and Mathematics, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia;
| |
Collapse
|
12
|
Habudele Z, Chen G, Qian SE, Vaughn MG, Zhang J, Lin H. High Dietary Intake of Iron Might Be Harmful to Atrial Fibrillation and Modified by Genetic Diversity: A Prospective Cohort Study. Nutrients 2024; 16:593. [PMID: 38474722 DOI: 10.3390/nu16050593] [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: 01/02/2024] [Revised: 02/03/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Some studies suggest an association between iron overload and cardiovascular diseases (CVDs). However, the relationship between dietary iron intake and atrial fibrillation (AF) remains uncertain, as does the role of genetic loci on this association. The study involved 179,565 participants from UK Biobank, tracking incident atrial fibrillation (AF) cases. Iron intake was categorized into low, moderate, and high groups based on dietary surveys conducted from 2009 to 2012. The Cox regression model was used to estimate the risk of AF in relation to iron intake, assessing the hazard ratio (HR) and 95% confidence interval (95% CI). It also examined the impact of 165 AF-related and 20 iron-related genetic variants on this association. Pathway enrichment analyses were performed using Metascape and FUMA. During a median follow-up period of 11.6 years, 6693 (3.97%) incident AF cases were recorded. A total of 35,874 (20.0%) participants had high iron intake. High iron intake was associated with increased risk of AF [HR: 1.13 (95% CI: 1.05, 1.22)] in a fully adjusted model. Importantly, there were 83 SNPs (11 iron-related SNPs) that could enhance the observed associations. These genes are mainly involved in cardiac development and cell signal transduction pathways. High dietary iron intake increases the risk of atrial fibrillation, especially when iron intake exceeds 16.95 mg. The association was particularly significant among the 83 SNPs associated with AF and iron, the individuals with these risk genes. Gene enrichment analysis revealed that these genes are significantly involved in cardiac development and cell signal transduction processes.
Collapse
Affiliation(s)
- Zierdi Habudele
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510275, China
| | - Ge Chen
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510275, China
| | - Samantha E Qian
- College of Arts and Sciences, Saint Louis University, St. Louis, MO 63108, USA
| | - Michael G Vaughn
- School of Social Work, Saint Louis University, St. Louis, MO 63103, USA
| | - Junguo Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510275, China
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
13
|
Singh AA, Shetty DK, Jacob AG, Bayraktar S, Sinha S. Understanding genomic medicine for thoracic aortic disease through the lens of induced pluripotent stem cells. Front Cardiovasc Med 2024; 11:1349548. [PMID: 38440211 PMCID: PMC10910110 DOI: 10.3389/fcvm.2024.1349548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/31/2024] [Indexed: 03/06/2024] Open
Abstract
Thoracic aortic disease (TAD) is often silent until a life-threatening complication occurs. However, genetic information can inform both identification and treatment at an early stage. Indeed, a diagnosis is important for personalised surveillance and intervention plans, as well as cascade screening of family members. Currently, only 20% of heritable TAD patients have a causative mutation identified and, consequently, further advances in genetic coverage are required to define the remaining molecular landscape. The rapid expansion of next generation sequencing technologies is providing a huge resource of genetic data, but a critical issue remains in functionally validating these findings. Induced pluripotent stem cells (iPSCs) are patient-derived, reprogrammed cell lines which allow mechanistic insights, complex modelling of genetic disease and a platform to study aortic genetic variants. This review will address the need for iPSCs as a frontline diagnostic tool to evaluate variants identified by genomic discovery studies and explore their evolving role in biological insight through to drug discovery.
Collapse
Affiliation(s)
| | | | | | | | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, United Kingdom
| |
Collapse
|
14
|
DeGroat W, Abdelhalim H, Patel K, Mendhe D, Zeeshan S, Ahmed Z. Discovering biomarkers associated and predicting cardiovascular disease with high accuracy using a novel nexus of machine learning techniques for precision medicine. Sci Rep 2024; 14:1. [PMID: 38167627 PMCID: PMC10762256 DOI: 10.1038/s41598-023-50600-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
Personalized interventions are deemed vital given the intricate characteristics, advancement, inherent genetic composition, and diversity of cardiovascular diseases (CVDs). The appropriate utilization of artificial intelligence (AI) and machine learning (ML) methodologies can yield novel understandings of CVDs, enabling improved personalized treatments through predictive analysis and deep phenotyping. In this study, we proposed and employed a novel approach combining traditional statistics and a nexus of cutting-edge AI/ML techniques to identify significant biomarkers for our predictive engine by analyzing the complete transcriptome of CVD patients. After robust gene expression data pre-processing, we utilized three statistical tests (Pearson correlation, Chi-square test, and ANOVA) to assess the differences in transcriptomic expression and clinical characteristics between healthy individuals and CVD patients. Next, the recursive feature elimination classifier assigned rankings to transcriptomic features based on their relation to the case-control variable. The top ten percent of commonly observed significant biomarkers were evaluated using four unique ML classifiers (Random Forest, Support Vector Machine, Xtreme Gradient Boosting Decision Trees, and k-Nearest Neighbors). After optimizing hyperparameters, the ensembled models, which were implemented using a soft voting classifier, accurately differentiated between patients and healthy individuals. We have uncovered 18 transcriptomic biomarkers that are highly significant in the CVD population that were used to predict disease with up to 96% accuracy. Additionally, we cross-validated our results with clinical records collected from patients in our cohort. The identified biomarkers served as potential indicators for early detection of CVDs. With its successful implementation, our newly developed predictive engine provides a valuable framework for identifying patients with CVDs based on their biomarker profiles.
Collapse
Affiliation(s)
- William DeGroat
- Health Care Policy and Aging Research, Rutgers Institute for Health, Rutgers University, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Habiba Abdelhalim
- Health Care Policy and Aging Research, Rutgers Institute for Health, Rutgers University, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Kush Patel
- Health Care Policy and Aging Research, Rutgers Institute for Health, Rutgers University, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Dinesh Mendhe
- Health Care Policy and Aging Research, Rutgers Institute for Health, Rutgers University, 112 Paterson St, New Brunswick, NJ, 08901, USA
| | - Saman Zeeshan
- Rutgers Cancer Institute of New Jersey, Rutgers University, 195 Little Albany St, New Brunswick, NJ, USA
| | - Zeeshan Ahmed
- Health Care Policy and Aging Research, Rutgers Institute for Health, Rutgers University, 112 Paterson St, New Brunswick, NJ, 08901, USA.
- Department of Medicine/Cardiovascular Disease and Hypertension, Robert Wood Johnson Medical School, Rutgers Biomedical and Health Sciences, 125 Paterson St, New Brunswick, NJ, USA.
| |
Collapse
|
15
|
Ahmed Z, Degroat W, Abdelhalim H, Zeeshan S, Fine D. Deciphering genomic signatures associating human dental oral craniofacial diseases with cardiovascular diseases using machine learning approaches. Clin Oral Investig 2024; 28:52. [PMID: 38163819 DOI: 10.1007/s00784-023-05406-3] [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: 06/29/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVES Periodontal diseases are chronic, inflammatory disorders that involve the destruction of supporting tissues surrounding the teeth which leads to permanent damage and substantially heightens systemic exposure. If left untreated, dental, oral, and craniofacial diseases (DOCs), especially periodontitis, can increase an individual's risk in developing complex traits including cardiovascular diseases (CVDs). In this study, we are focused on systematically investigating causality between periodontitis with CVDs with the application of artificial intelligence (AI), machine learning (ML) algorithms, and state-of-the-art bioinformatics approaches using RNA-seq-driven gene expression data of CVD patients. MATERIALS AND METHODS In this study, we built a cohort of CVD patients, collected their blood samples, and performed RNA-seq and gene expression analysis to generate transcriptomic profiles. We proposed a nexus of AI/ML approaches for the identification of significant biomarkers, and predictive analysis. We implemented recursive feature elimination, Pearson correlation, chi-square, and analysis of variance to detect significant biomarkers, and utilized random forest and support vector machines for predictive analysis. RESULTS Our AI/ML analyses have led us to the preliminary conclusion that GAS5, GPX1, HLA-B, and SNHG6 are the potential gene markers that can be used to explain the causal relationship between periodontitis and CVDs. CONCLUSIONS CVDs are relatively common in patients with periodontal disease, and an increased risk of CVD is associated with periodontal disease independent of gender. Genetic susceptibility contributing to periodontitis and CVDs have been suggested to some extent, based on the similar degree of heritability shared between both complex diseases.
Collapse
Affiliation(s)
- Zeeshan Ahmed
- Department of Medicine/Cardiovascular Disease and Hypertension, Robert Wood Johnson Medical School, Rutgers Biomedical and Health Sciences, 125 Paterson St, New Brunswick, NJ, USA.
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA.
| | - William Degroat
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Habiba Abdelhalim
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson St, New Brunswick, NJ, USA
| | - Saman Zeeshan
- Department of Radiation Oncology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO, USA
| | - Daniel Fine
- Department of Oral Biology, Rutgers School of Dental Medicine, 110 Bergen Street, Newark, NJ, USA
| |
Collapse
|
16
|
Wang X, Qi M, Zhang H, Yang Y, Zhao H. Genome-wide association and Mendelian randomization analysis provide insights into the shared genetic architecture between high-dimensional electrocardiographic features and ischemic heart disease. Hum Genet 2024; 143:49-58. [PMID: 38180560 DOI: 10.1007/s00439-023-02614-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/27/2023] [Indexed: 01/06/2024]
Abstract
Observational studies have revealed that ischemic heart disease (IHD) has a unique manifestation on electrocardiographic (ECG). However, the genetic relationships between IHD and ECG remain unclear. We took 12-lead ECG as phenotypes to conduct genome-wide association studies (GWAS) for 41,960 samples from UK-Biobank (UKB). By leveraging large-scale GWAS summary of ECG and IHD (downloaded from FinnGen database), we performed LD score regression (LDSC), Mendelian randomization (MR), and polygenic risk score (PRS) regression to explore genetic relationships between IHD and ECG. Finally, we constructed an XGBoost model to predict IHD by integrating PRS and ECG. The GWAS identified 114 independent SNPs significantly (P value < 5 × 10-8/800, where 800 denotes the number of ECG features) associated with ECG. LDSC analysis indicated significant (P value < 0.05) genetic correlations between 39 ECG features and IHD. MR analysis performed by five approaches showed a putative causal effect of IHD on four S wave related ECG features at lead III. Integrating PRS for these ECG features with age and gender, the XGBoost model achieved Area Under Curve (AUC) 0.72 in predicting IHD. Here, we provide genetic evidence supporting S wave related ECG features at lead III to monitor the IHD risk, and open up a unique approach to integrate ECG with genetic factors for pre-warning IHD.
Collapse
Affiliation(s)
- Xinfeng Wang
- College of Computer Science and Engineering, Jishou University, Jishou, China
- Department of Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang West Road, Guangzhou, 500001, People's Republic of China
- Key Laboratory of Machine Intelligence and Advanced Computing of MOE, Guangzhou, China
| | - Mengling Qi
- Department of Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang West Road, Guangzhou, 500001, People's Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China
| | - Haoyang Zhang
- School of Computer Science and Engineering, Sun Yat-Sen University, Guangzhou, China
| | - Yuedong Yang
- School of Computer Science and Engineering, Sun Yat-Sen University, Guangzhou, China
| | - Huiying Zhao
- Department of Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang West Road, Guangzhou, 500001, People's Republic of China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, China.
| |
Collapse
|
17
|
Popa-Fotea NM, Oprescu N, Scafa-Udriste A, Micheu MM. Impact of rs1805127 and rs55742440 Variants on Atrial Remodeling in Hypertrophic Cardiomyopathy Patients with Atrial Fibrillation: A Romanian Cohort Study. Int J Mol Sci 2023; 24:17244. [PMID: 38139087 PMCID: PMC10743528 DOI: 10.3390/ijms242417244] [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: 10/11/2023] [Revised: 11/25/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Atrial fibrillation (AFib) is characterized by a complex genetic component. We aimed to investigate the association between variations in genes related to cardiac ion handling and AFib in a cohort of Romanian patients with hypertrophic cardiomyopathy (HCM). Forty-five unrelated probands with HCM were genotyped by targeted next-generation sequencing (NGS) for 24 genes associated with cardiac ion homeostasis. Subsequently, the study cohort was divided into two groups based on the presence (AFib+) or absence (AFiB-) of AFib detected during ECG monitoring. We identified two polymorphisms (rs1805127 located in KCNE1 and rs55742440 located in SCN1B) linked to AFib susceptibility. In AFib+, rs1805127 was associated with increased indexed left atrial (LA) maximal volume (LAVmax) (58.42 ± 21 mL/m2 vs. 32.54 ± 6.47 mL/m2, p < 0.001) and impaired LA strain reservoir (LASr) (13.3 ± 7.5% vs. 24.4 ± 6.8%, p < 0.05) compared to those without respective variants. The rs55742440 allele was less frequent in patients with AFib+ (12 out of 25, 48%) compared to those without arrhythmia (15 out of 20, 75%, p = 0.05). Also, AFib+ rs55742440 carriers had significantly lower LAVmax compared to those who were genotype negative. Among patients with HCM and AFib+, the rs1805127 variant was accompanied by pronounced LA remodeling, whereas rs55742440's presence was related to a milder LA enlargement.
Collapse
Affiliation(s)
- Nicoleta-Monica Popa-Fotea
- Department 4—Cardio-Thoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania;
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Calea Floreasca 8, 014461 Bucharest, Romania;
| | - Nicoleta Oprescu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Calea Floreasca 8, 014461 Bucharest, Romania;
| | - Alexandru Scafa-Udriste
- Department 4—Cardio-Thoracic Pathology, University of Medicine and Pharmacy Carol Davila, Eroii Sanitari Bvd. 8, 050474 Bucharest, Romania;
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Calea Floreasca 8, 014461 Bucharest, Romania;
| | - Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Calea Floreasca 8, 014461 Bucharest, Romania;
| |
Collapse
|
18
|
Mhatre I, Abdelhalim H, Degroat W, Ashok S, Liang BT, Ahmed Z. Functional mutation, splice, distribution, and divergence analysis of impactful genes associated with heart failure and other cardiovascular diseases. Sci Rep 2023; 13:16769. [PMID: 37798313 PMCID: PMC10556087 DOI: 10.1038/s41598-023-44127-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 10/04/2023] [Indexed: 10/07/2023] Open
Abstract
Cardiovascular disease (CVD) is caused by a multitude of complex and largely heritable conditions. Identifying key genes and understanding their susceptibility to CVD in the human genome can assist in early diagnosis and personalized treatment of the relevant patients. Heart failure (HF) is among those CVD phenotypes that has a high rate of mortality. In this study, we investigated genes primarily associated with HF and other CVDs. Achieving the goals of this study, we built a cohort of thirty-five consented patients, and sequenced their serum-based samples. We have generated and processed whole genome sequence (WGS) data, and performed functional mutation, splice, variant distribution, and divergence analysis to understand the relationships between each mutation type and its impact. Our variant and prevalence analysis found FLNA, CST3, LGALS3, and HBA1 linked to many enrichment pathways. Functional mutation analysis uncovered ACE, MME, LGALS3, NR3C2, PIK3C2A, CALD1, TEK, and TRPV1 to be notable and potentially significant genes. We discovered intron, 5' Flank, 3' UTR, and 3' Flank mutations to be the most common among HF and other CVD genes. Missense mutations were less common among HF and other CVD genes but had more of a functional impact. We reported HBA1, FADD, NPPC, ADRB2, ADBR1, MYH6, and PLN to be consequential based on our divergence analysis.
Collapse
Affiliation(s)
- Ishani Mhatre
- Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Habiba Abdelhalim
- Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - William Degroat
- Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Shreya Ashok
- Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA
| | - Bruce T Liang
- Pat and Jim Calhoun Cardiology Center, UConn Health, 263 Farmington Ave, Farmington, CT, USA
- UConn School of Medicine, University of Connecticut, 263 Farmington Ave, Farmington, CT, USA
| | - Zeeshan Ahmed
- Institute for Health, Health Care Policy and Aging Research, Rutgers University, 112 Paterson Street, New Brunswick, NJ, 08901, USA.
- Department of Genetics and Genome Sciences, UConn Health, 400 Farmington Ave, Farmington, CT, USA.
- Department of Medicine/Cardiovascular Disease and Hypertension, Robert Wood Johnson Medical School, Rutgers Biomedical and Health Sciences, 125 Paterson St, New Brunswick, NJ, USA.
| |
Collapse
|
19
|
Liantonio A, Bertini M, Mele A, Balla C, Dinoi G, Selvatici R, Mele M, De Luca A, Gualandi F, Imbrici P. Brugada Syndrome: More than a Monogenic Channelopathy. Biomedicines 2023; 11:2297. [PMID: 37626795 PMCID: PMC10452102 DOI: 10.3390/biomedicines11082297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Brugada syndrome (BrS) is an inherited cardiac channelopathy first diagnosed in 1992 but still considered a challenging disease in terms of diagnosis, arrhythmia risk prediction, pathophysiology and management. Despite about 20% of individuals carrying pathogenic variants in the SCN5A gene, the identification of a polygenic origin for BrS and the potential role of common genetic variants provide the basis for applying polygenic risk scores for individual risk prediction. The pathophysiological mechanisms are still unclear, and the initial thinking of this syndrome as a primary electrical disease is evolving towards a partly structural disease. This review focuses on the main scientific advancements in the identification of biomarkers for diagnosis, risk stratification, pathophysiology and therapy of BrS. A comprehensive model that integrates clinical and genetic factors, comorbidities, age and gender, and perhaps environmental influences may provide the opportunity to enhance patients' quality of life and improve the therapeutic approach.
Collapse
Affiliation(s)
- Antonella Liantonio
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.L.); (A.M.); (G.D.); (M.M.); (A.D.L.)
| | - Matteo Bertini
- Cardiological Center, Sant’Anna University Hospital of Ferrara, 44121 Ferrara, Italy; (M.B.); (C.B.)
| | - Antonietta Mele
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.L.); (A.M.); (G.D.); (M.M.); (A.D.L.)
| | - Cristina Balla
- Cardiological Center, Sant’Anna University Hospital of Ferrara, 44121 Ferrara, Italy; (M.B.); (C.B.)
| | - Giorgia Dinoi
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.L.); (A.M.); (G.D.); (M.M.); (A.D.L.)
| | - Rita Selvatici
- Medical Genetics Unit, Department of Mother and Child, Sant’Anna University Hospital of Ferrara, 44121 Ferrara, Italy;
| | - Marco Mele
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.L.); (A.M.); (G.D.); (M.M.); (A.D.L.)
- Cardiothoracic Department, Policlinico Riuniti Foggia, 71122 Foggia, Italy
| | - Annamaria De Luca
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.L.); (A.M.); (G.D.); (M.M.); (A.D.L.)
| | - Francesca Gualandi
- Medical Genetics Unit, Department of Mother and Child, Sant’Anna University Hospital of Ferrara, 44121 Ferrara, Italy;
| | - Paola Imbrici
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (A.L.); (A.M.); (G.D.); (M.M.); (A.D.L.)
| |
Collapse
|
20
|
Darkow E, Yusuf D, Rajamani S, Backofen R, Kohl P, Ravens U, Peyronnet R. Meta-Analysis of Mechano-Sensitive Ion Channels in Human Hearts: Chamber- and Disease-Preferential mRNA Expression. Int J Mol Sci 2023; 24:10961. [PMID: 37446137 DOI: 10.3390/ijms241310961] [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: 05/07/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
The cardiac cell mechanical environment changes on a beat-by-beat basis as well as in the course of various cardiac diseases. Cells sense and respond to mechanical cues via specialized mechano-sensors initiating adaptive signaling cascades. With the aim of revealing new candidates underlying mechano-transduction relevant to cardiac diseases, we investigated mechano-sensitive ion channels (MSC) in human hearts for their chamber- and disease-preferential mRNA expression. Based on a meta-analysis of RNA sequencing studies, we compared the mRNA expression levels of MSC in human atrial and ventricular tissue samples from transplant donor hearts (no cardiac disease), and from patients in sinus rhythm (underlying diseases: heart failure, coronary artery disease, heart valve disease) or with atrial fibrillation. Our results suggest that a number of MSC genes are expressed chamber preferentially, e.g., CHRNE in the atria (compared to the ventricles), TRPV4 in the right atrium (compared to the left atrium), CACNA1B and KCNMB1 in the left atrium (compared to the right atrium), as well as KCNK2 and KCNJ2 in ventricles (compared to the atria). Furthermore, 15 MSC genes are differentially expressed in cardiac disease, out of which SCN9A (lower expressed in heart failure compared to donor tissue) and KCNQ5 (lower expressed in atrial fibrillation compared to sinus rhythm) show a more than twofold difference, indicative of possible functional relevance. Thus, we provide an overview of cardiac MSC mRNA expression in the four cardiac chambers from patients with different cardiac diseases. We suggest that the observed differences in MSC mRNA expression may identify candidates involved in altered mechano-transduction in the respective diseases.
Collapse
Affiliation(s)
- Elisa Darkow
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg∙Bad Krozingen, 79110 Freiburg im Breisgau, Germany
- Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Dilmurat Yusuf
- Bioinformatics Group, Department of Computer Science, University of Freiburg, 79110 Freiburg im Breisgau, Germany
| | - Sridharan Rajamani
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Inc., South San Francisco, CA 91320, USA
| | - Rolf Backofen
- Bioinformatics Group, Department of Computer Science, University of Freiburg, 79110 Freiburg im Breisgau, Germany
- Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg∙Bad Krozingen, 79110 Freiburg im Breisgau, Germany
- Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany
- Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Ursula Ravens
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg∙Bad Krozingen, 79110 Freiburg im Breisgau, Germany
- Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg∙Bad Krozingen, 79110 Freiburg im Breisgau, Germany
- Medical Center and Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany
| |
Collapse
|