1
|
Lasko TA, Stead WW, Still JM, Li TZ, Kammer M, Barbero-Mota M, Strobl EV, Landman BA, Maldonado F. Unsupervised discovery of clinical disease signatures using probabilistic independence. J Biomed Inform 2025; 166:104837. [PMID: 40280380 DOI: 10.1016/j.jbi.2025.104837] [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: 11/25/2024] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
OBJECTIVE This study uses probabilistic independence to disentangle patient-specific sources of disease and their signatures in Electronic Health Record (EHR) data. MATERIALS AND METHODS We model a disease source as an unobserved root node in the causal graph of observed EHR variables (laboratory test results, medication exposures, billing codes, and demographics), and a signature as the set of downstream effects that a given source has on those observed variables. We used probabilistic independence to infer 2000 sources and their signatures from 9195 variables in 630,000 cross-sectional training instances sampled at random times from 269,099 longitudinal patient records. We evaluated the learned sources by using them to infer and explain the causes of benign vs. malignant pulmonary nodules in 13,252 records, comparing the inferred causes to an external reference list and other medical literature. We compared models trained by three different algorithms and used corresponding models trained directly from the observed variables as baselines. RESULTS The model recovered 92% of malignant and 30% of benign causes in the reference standard. Of the top 20 inferred causes of malignancy, 14 were not listed in the reference standard, but had supporting evidence in the literature, as did 11 of the top 20 inferred causes of benign nodules. The model decomposed listed malignant causes by an average factor of 5.5 and benign causes by 4.1, with most stratifying by disease course or treatment regimen. Predictive accuracy of causal predictive models trained on source expressions (Random Forest AUC 0.788) was similar to (p = 0.058) their associational baselines (0.738). DISCUSSION Most of the unrecovered causes were due to the rarity of the condition or lack of sufficient detail in the input data. Surprisingly, the causal model found many patients with apparently undiagnosed cancer as the source of the malignant nodules. Causal model AUC also suggests that some sources remained undiscovered in this cohort. CONCLUSION These promising results demonstrate the potential of using probabilistic independence to disentangle complex clinical signatures from noisy, asynchronous, and incomplete EHR data that represent the confluence of multiple simultaneous conditions, and to identify patient-specific causes that support precise treatment decisions.
Collapse
Affiliation(s)
- Thomas A Lasko
- Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA; Vanderbilt University, 2301 Vanderbilt Pl, Nashville, TN 37235, USA.
| | - William W Stead
- Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA
| | - John M Still
- Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA
| | - Thomas Z Li
- Vanderbilt University, 2301 Vanderbilt Pl, Nashville, TN 37235, USA
| | - Michael Kammer
- Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA
| | - Marco Barbero-Mota
- Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA
| | - Eric V Strobl
- Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA; University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260, USA
| | - Bennett A Landman
- Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA; Vanderbilt University, 2301 Vanderbilt Pl, Nashville, TN 37235, USA
| | - Fabien Maldonado
- Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA
| |
Collapse
|
2
|
Nogueira M, Sanchez-Martinez S, Piella G, De Craene M, Yagüe C, Marti-Castellote PM, Bonet M, Oladapo OT, Bijnens B. Labour monitoring and decision support: a machine-learning-based paradigm. Front Glob Womens Health 2025; 6:1368575. [PMID: 40309718 PMCID: PMC12040997 DOI: 10.3389/fgwh.2025.1368575] [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: 01/10/2024] [Accepted: 02/17/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction A machine-learning-based paradigm, combining unsupervised and supervised components, is proposed for the problem of real-time monitoring and decision support during labour, addressing the limitations of current state-of-the-art approaches, such as the partograph or purely supervised models. Methods The proposed approach is illustrated with World Health Organisation's Better Outcomes in Labour Difficulty (BOLD) prospective cohort study data, including 9,995 women admitted for labour in 2014-2015 in thirteen major regional health care facilities across Nigeria and Uganda. Unsupervised dimensionality reduction is used to map complex labour data to a visually intuitive space. In this space, an ongoing labour trajectory can be compared to those of a historical cohort of women with similar characteristics and known outcomes-this information can be used to estimate personalised "healthy" trajectory references (and alert the healthcare provider to significant deviations), as well as draw attention to high incidences of different interventions/adverse outcomes among similar labours. To evaluate the proposed approach, the predictive value of simple risk scores quantifying deviation from normal progress and incidence of complications among similar labours is assessed in a caesarean section prediction context and compared to that of the partograph and state-of-the-art supervised machine-learning models. Results Considering all women, our predictors yielded sensitivity and specificity of ∼0.70. It was observed that this predictive performance could increase or decrease when looking at different subgroups. Discussion With a simple implementation, our approach outperforms the partograph and matches the performance of state-of-the-art supervised models, while offering superior flexibility and interpretability as a real-time monitoring and decision-support solution.
Collapse
Affiliation(s)
- Mariana Nogueira
- Department of Engineering, Universitat Pompeu Fabra, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Sergio Sanchez-Martinez
- Department of Engineering, Universitat Pompeu Fabra, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Gemma Piella
- Department of Engineering, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Carlos Yagüe
- Department of Engineering, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Mercedes Bonet
- UNDP/UNFPA/UNICEF/WHO/World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), Department of Sexual and Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Olufemi T. Oladapo
- UNDP/UNFPA/UNICEF/WHO/World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), Department of Sexual and Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Bart Bijnens
- Department of Engineering, Universitat Pompeu Fabra, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
- ICREA, Barcelona, Spain
| |
Collapse
|
3
|
Ji Y, Cutiongco MFA, Jensen BS, Yuan K. Generating realistic single-cell images from CellProfiler representations. Med Image Anal 2025; 103:103574. [PMID: 40393380 DOI: 10.1016/j.media.2025.103574] [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: 02/01/2024] [Revised: 12/07/2024] [Accepted: 03/28/2025] [Indexed: 05/22/2025]
Abstract
High-throughput imaging techniques acquire large amounts of images efficiently. These images contain rich biological information including cellular processes. A common method to analyse them is to encode them into quantitative representation vectors. Generally, there are two ways to extract cell biological information into representations, hand-crafted and machine-learning. Although representations obtained from machine learning models often demonstrate commendable reconstruction performance, they lack biological interpretability. In contrast, hand-crafted representations have clear biological meanings, making them easily interpretable. However, the capability of hand-crafted representations to generate realistic images remains uncertain. In this work, we propose a CellProfiler to image (CP2Image) model capable of directly generating realistic cell images from CellProfiler representations. The proposed model is demonstrated to be robust to different architectures, including ResNet, InceptionNet and Transformer. We also show that the biological information is well preserved during the generation process. The changes in certain CellProfiler features will reflect the corresponding changes in the generated single-cell images. In addition, the CP2Image model can generate conditional phenotypes, which will ultimately help diagnostics and drug screening.
Collapse
Affiliation(s)
- Yanni Ji
- School of Computing Science, University of Glasgow, Glasgow, G12 8RZ, Scotland, UK.
| | - Marie F A Cutiongco
- James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, Scotland, UK.
| | - Bjørn Sand Jensen
- School of Computing Science, University of Glasgow, Glasgow, G12 8RZ, Scotland, UK; Technically University of Denmark, DTU Compute, Kgs, Lyngby, Denmark.
| | - Ke Yuan
- School of Computing Science, University of Glasgow, Glasgow, G12 8RZ, Scotland, UK; School of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, Scotland, UK; Cancer Research UK Beatson Institute, Glasgow, G61 1BD, Scotland, UK.
| |
Collapse
|
4
|
Jin K, Yu H, Wei W, Grzybowski A. Editorial: Personalized medicine of diabetes retinopathy: from bench to bedside. Front Endocrinol (Lausanne) 2025; 16:1597332. [PMID: 40235660 PMCID: PMC11997570 DOI: 10.3389/fendo.2025.1597332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025] Open
Affiliation(s)
- Kai Jin
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Engineering Institute on Eye Diseases, Zhejiang University, Hangzhou, China
| | - Honghua Yu
- Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People’s Hospital, Southern Medical University, Guangzhou, China
| | - Wenbin Wei
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Andrzej Grzybowski
- Institute for Research in Ophthalmology, Foundation for Ophthalmology Development, Poznan, Poland
- Department of Ophthalmology, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|
5
|
Loughrey CF, Maguire S, Dłotko P, Bai L, Orr N, Jurek-Loughrey A. A novel method for subgroup discovery in precision medicine based on topological data analysis. BMC Med Inform Decis Mak 2025; 25:139. [PMID: 40102808 PMCID: PMC11921513 DOI: 10.1186/s12911-025-02852-9] [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] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/03/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND The Mapper algorithm is a data mining topological tool that can help us to obtain higher level understanding of disease by visualising the structure of patient data as a similarity graph. It has been successfully applied for exploratory analysis of cancer data in the past, delivering several significant subgroup discoveries. Using the Mapper algorithm in practice requires setting up multiple parameters. The graph then needs to be manually analysed according to a research question at hand. It has been highlighted in the literature that Mapper's parameters have significant impact on the output graph shape and there is no established way to select their optimal values. Hence while using the Mapper algorithm, different parameter values and consequently different output graphs need to be studied. This prevents routine application of the Mapper algorithm in real world settings. METHODS We propose a new algorithm for subgroup discovery within the Mapper graph. We refer to the task as hotspot detection as it is designed to identify homogenous and geometrically compact subsets of patients, which are distinct with respect to their clinical or molecular profiles (e.g. survival). Furthermore, we propose to include the existence of a hotspot as a criterion while searching the parameter space, addressing one of the key limitations of the Mapper algorithm (i.e. parameter selection). RESULTS Two experiments were performed to demonstrate the efficacy of the algorithm, including an artificial hotspot in the Two Circles dataset and a real world case study of subgroup discovery in oestrogen receptor-positive breast cancer. Our hotspot detection algorithm successfully identified graphs containing homogenous communities of nodes within the Two Circles dataset. When applied to gene expression data of ER+ breast cancer patients, appropriate parameters were identified to generate a Mapper graph revealing a hotspot of ER+ patients with poor prognosis and characteristic patterns of gene expression. This was subsequently confirmed in an independent breast cancer dataset. CONCLUSIONS Our proposed method can be effectively applied for subgroup discovery with pathology data. It allows us to find optimal parameters of the Mapper algorithm, bridging the gap between its potential and the translational research.
Collapse
Affiliation(s)
- Ciara F Loughrey
- School of Electronics, Electrical Engineering and Computer Science, Queen's University Belfast, Belfast, UK
| | - Sarah Maguire
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Paweł Dłotko
- Dioscuri Centre in Topological Data Analysis, Mathematical Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Lu Bai
- School of Electronics, Electrical Engineering and Computer Science, Queen's University Belfast, Belfast, UK
| | - Nick Orr
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Anna Jurek-Loughrey
- School of Electronics, Electrical Engineering and Computer Science, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
6
|
Lambert J, Leutenegger AL, Baudot A, Jannot AS. Improving patient clustering by incorporating structured variable label relationships in similarity measures. BMC Med Res Methodol 2025; 25:72. [PMID: 40089699 PMCID: PMC11910865 DOI: 10.1186/s12874-025-02459-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 01/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Patient stratification is the cornerstone of numerous health investigations, serving to enhance the estimation of treatment efficacy and facilitating patient matching. To stratify patients, similarity measures between patients can be computed from clinical variables contained in medical health records. These variables have both values and labels structured in ontologies or other classification systems. The relevance of considering variable label relationships in the computation of patient similarity measures has been poorly studied. OBJECTIVE We adapt and evaluate several weighted versions of the Cosine similarity in order to consider structured label relationships to compute patient similarities from a medico-administrative database. MATERIALS AND METHODS As a use case, we clustered patients aged 60 years from their annual medicine reimbursements contained in the Échantillon Généraliste des Bénéficiaires, a random sample of a French medico-administrative database. We used four patient similarity measures: the standard Cosine similarity, a weighted Cosine similarity measure that includes variable frequencies and two weighted Cosine similarity measures that consider variable label relationships. We construct patient networks from each similarity measure and identify clusters of patients using the Markov Cluster algorithm. We evaluate the performance of the different similarity measures with enrichment tests based on patient diagnoses. RESULTS The weighted similarity measures that include structured variable label relationships perform better to identify similar patients. Indeed, using these weighted measures, we identify more clusters associated with different diagnose enrichment. Importantly, the enrichment tests provide clinically interpretable insights into these patient clusters. CONCLUSION Considering label relationships when computing patient similarities improves stratification of patients regarding their health status.
Collapse
Affiliation(s)
- Judith Lambert
- Sorbonne Université, Université Paris Cité, INSERM, Centre de Recherche des Cordeliers, Paris, F-75006, France.
- HeKA, Inria Paris, Paris, F-75015, France.
- Aix Marseille Univ, INSERM, MMG, Marseille, UMR1251, France.
| | | | - Anaïs Baudot
- Aix Marseille Univ, INSERM, MMG, Marseille, UMR1251, France
- CNRS, Marseille, France
- Barcelona Supercomputing Center, Barcelona, Spain
| | - Anne-Sophie Jannot
- HeKA, Inria Paris, Paris, F-75015, France
- Université Paris Cité, Sorbonne Université, INSERM, Centre de Recherche des Cordeliers, F-75006, Paris, France
- French National Rare Disease Registry (BNDMR), Greater Paris University Hospitals (AP-HP), Paris, France
| |
Collapse
|
7
|
Bazzazzadehgan S, Shariat-Madar Z, Mahdi F. Distinct Roles of Common Genetic Variants and Their Contributions to Diabetes: MODY and Uncontrolled T2DM. Biomolecules 2025; 15:414. [PMID: 40149950 PMCID: PMC11940602 DOI: 10.3390/biom15030414] [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] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/26/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) encompasses a range of clinical manifestations, with uncontrolled diabetes leading to progressive or irreversible damage to various organs. Numerous genes associated with monogenic diabetes, exhibiting classical patterns of inheritance (autosomal dominant or recessive), have been identified. Additionally, genes involved in complex diabetes, which interact with environmental factors to trigger the disease, have also been discovered. These genetic findings have raised hopes that genetic testing could enhance diagnostics, disease surveillance, treatment selection, and family counseling. However, the accurate interpretation of genetic data remains a significant challenge, as variants may not always be definitively classified as either benign or pathogenic. Research to date, however, indicates that periodic reevaluation of genetic variants in diabetes has led to more consistent findings, with biases being steadily eliminated. This has improved the interpretation of variants across diverse ethnicities. Clinical studies suggest that genetic risk information may motivate patients to adopt behaviors that promote the prevention or management of T2DM. Given that the clinical features of certain monogenic diabetes types overlap with T2DM, and considering the significant role of genetic variants in diabetes, healthcare providers caring for prediabetic patients should consider genetic testing as part of the diagnostic process. This review summarizes current knowledge of the most common genetic variants associated with T2DM, explores novel therapeutic targets, and discusses recent advancements in the pharmaceutical management of uncontrolled T2DM.
Collapse
Affiliation(s)
- Shadi Bazzazzadehgan
- Department of Pharmacy Administration, School of Pharmacy, University of Mississippi, University, MS 38677, USA;
| | - Zia Shariat-Madar
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA;
| | - Fakhri Mahdi
- Division of Pharmacology, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA;
| |
Collapse
|
8
|
Tian X, Wang L, Zhong L, Zhang K, Ge X, Luo Z, Zhai X, Liu S. The research progress and future directions in the pathophysiological mechanisms of type 2 diabetes mellitus from the perspective of precision medicine. Front Med (Lausanne) 2025; 12:1555077. [PMID: 40109716 PMCID: PMC11919862 DOI: 10.3389/fmed.2025.1555077] [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: 01/03/2025] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex metabolic disorder characterized by pathophysiological mechanisms such as insulin resistance and β-cell dysfunction. Recent advancements in T2DM research have unveiled intricate multi-level regulatory networks and contributing factors underlying this disease. The emergence of precision medicine has introduced new perspectives and methodologies for understanding T2DM pathophysiology. A recent study found that personalized treatment based on genetic, metabolic, and microbiome data can improve the management of T2DM by more than 30%. This perspective aims to summarize the progress in T2DM pathophysiological research from the past 5 years and to outline potential directions for future studies within the framework of precision medicine. T2DM develops through the interplay of factors such as gut microbiota, genetic and epigenetic modifications, metabolic processes, mitophagy, NK cell activity, and environmental influences. Future research should focus on understanding insulin resistance, β-cell dysfunction, interactions between gut microbiota and their metabolites, and the regulatory roles of miRNA and genes. By leveraging artificial intelligence and integrating data from genomics, epigenomics, metabolomics, and microbiomics, researchers can gain deeper insights into the pathophysiological mechanisms and heterogeneity of T2DM. Additionally, exploring the combined effects and interactions of these factors may pave the way for more effective prevention strategies and personalized treatments for T2DM.
Collapse
Affiliation(s)
- Xinyi Tian
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liuqing Wang
- Institute of Chinese Medical History and Literatures, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liuting Zhong
- First School of Clinical Medicine, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Kaiqi Zhang
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolei Ge
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhengrong Luo
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xu Zhai
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shaoyan Liu
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Lindsay‐McGee V, Massey C, Li YT, Clark EL, Psifidi A, Piercy RJ. Characterisation of phenotypic patterns in equine exercise-associated myopathies. Equine Vet J 2025; 57:347-361. [PMID: 38965932 PMCID: PMC11807944 DOI: 10.1111/evj.14128] [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: 10/09/2023] [Accepted: 06/05/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Equine exercise-associated myopathies are prevalent, clinically heterogeneous, generally idiopathic disorders characterised by episodes of myofibre damage that occur in association with exercise. Episodes are intermittent and vary within and between affected horses and across breeds. The aetiopathogenesis is often unclear; there might be multiple causes. Poor phenotypic characterisation hinders genetic and other disease analyses. OBJECTIVES The aim of this study was to characterise phenotypic patterns across exercise-associated myopathies in horses. STUDY DESIGN Historical cross-sectional study, with subsequent masked case-control validation study. METHODS Historical clinical and histological features from muscle samples (n = 109) were used for k-means clustering and validated using principal components analysis and hierarchical clustering. For further validation, a blinded histological study (69 horses) was conducted comparing two phenotypic groups with selected controls and horses with histopathological features characterised by myofibrillar disruption. RESULTS We identified two distinct broad phenotypes: a non-classic exercise-associated myopathy syndrome (EAMS) subtype was associated with practitioner-described signs of apparent muscle pain (p < 0.001), reluctance to move (10.85, p = 0.001), abnormal gait (p < 0.001), ataxia (p = 0.001) and paresis (p = 0.001); while a non-specific classic RER subtype was not uniquely associated with any particular variables. No histological differences were identified between subtypes in the validation study, and no identifying histopathological features for other equine myopathies identified in either subtype. MAIN LIMITATIONS Lack of an independent validation population; small sample size of smaller identified subtypes; lack of positive control myofibrillar myopathy cases; case descriptions derived from multiple independent and unblinded practitioners. CONCLUSIONS This is the first study using computational clustering methods to identify phenotypic patterns in equine exercise-associated myopathies, and suggests that differences in patterns of presenting clinical signs support multiple disease subtypes, with EAMS a novel subtype not previously described. Routine muscle histopathology was not helpful in sub-categorising the phenotypes in our population.
Collapse
Affiliation(s)
- Victoria Lindsay‐McGee
- Department of Clinical Sciences and ServicesRoyal Veterinary CollegeLondonUK
- Present address:
Royal (Dick) School of Veterinary StudiesUniversity of EdinburghEdinburghUK
| | - Claire Massey
- Department of Clinical Sciences and ServicesRoyal Veterinary CollegeLondonUK
| | - Ying Ting Li
- Department of Clinical Sciences and ServicesRoyal Veterinary CollegeLondonUK
| | - Emily L. Clark
- The Roslin Institute, University of EdinburghEdinburghUK
| | - Androniki Psifidi
- Department of Clinical Sciences and ServicesRoyal Veterinary CollegeLondonUK
- The Roslin Institute, University of EdinburghEdinburghUK
| | - Richard J. Piercy
- Department of Clinical Sciences and ServicesRoyal Veterinary CollegeLondonUK
| |
Collapse
|
10
|
Agostoni P, Chiesa M, Salvioni E, Emdin M, Piepoli M, Sinagra G, Senni M, Bonomi A, Adamopoulos S, Miliopoulos D, Mapelli M, Campodonico J, Attanasio U, Apostolo A, Pestrin E, Rossoni A, Magrì D, Paolillo S, Corrà U, Raimondo R, Cittadini A, Iorio A, Salzano A, Lagioia R, Vignati C, Badagliacca R, Filardi PP, Correale M, Perna E, Metra M, Cattadori G, Guazzi M, Limongelli G, Parati G, De Martino F, Matassini MV, Bandera F, Bussotti M, Re F, Lombardi CM, Scardovi AB, Sciomer S, Passantino A, Santolamazza C, Girola D, Passino C, Karsten M, Nodari S, Pompilio G. The chronic heart failure evolutions: Different fates and routes. ESC Heart Fail 2025; 12:418-433. [PMID: 39318188 PMCID: PMC11769638 DOI: 10.1002/ehf2.14966] [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: 03/13/2024] [Revised: 05/09/2024] [Accepted: 06/24/2024] [Indexed: 09/26/2024] Open
Abstract
AIMS Individual prognostic assessment and disease evolution pathways are undefined in chronic heart failure (HF). The application of unsupervised learning methodologies could help to identify patient phenotypes and the progression in each phenotype as well as to assess adverse event risk. METHODS AND RESULTS From a bulk of 7948 HF patients included in the MECKI registry, we selected patients with a minimum 2-year follow-up. We implemented a topological data analysis (TDA), based on 43 variables derived from clinical, biochemical, cardiac ultrasound, and exercise evaluations, to identify several patients' clusters. Thereafter, we used the trajectory analysis to describe the evolution of HF states, which is able to identify bifurcation points, characterized by different follow-up paths, as well as specific end-stages conditions of the disease. Finally, we conducted a 5-year survival analysis (composite of cardiovascular death, left ventricular assist device, or urgent heart transplant). Findings were validated on internal (n = 527) and external (n = 777) populations. We analyzed 4876 patients (age = 63 [53-71], male gender n = 3973 (81.5%), NYHA class I-II n = 3576 (73.3%), III-IV n = 1300 (26.7%), LVEF = 33 [25.5-39.9], atrial fibrillation n = 791 (16.2%), peak VO2% pred = 54.8 [43.8-67.2]), with a minimum 2-year follow-up. Nineteen patient clusters were identified by TDA. Trajectory analysis revealed a path characterized by 3 bifurcation and 4 end-stage points. Clusters survival rate varied from 44% to 100% at 2 years and from 20% to 100% at 5 years, respectively. The event frequency at 5-year follow-up for each study cohort cluster was successfully compared with those in the validation cohorts (R = 0.94 and R = 0.84, P < 0.001, for internal and external cohort, respectively). Finally, we conducted a 5-year survival analysis (composite of cardiovascular death, left ventricular assist device, or urgent heart transplant observed in 22% of cases). CONCLUSIONS Each HF phenotype has a specific disease progression and prognosis. These findings allow to individualize HF patient evolutions and to tailor assessment.
Collapse
Affiliation(s)
- Piergiuseppe Agostoni
- Centro Cardiologico Monzino, IRCCsMilanItaly
- Department of Clinical Sciences and Community Health, Section of CardiologyUniversity of MilanMilanItaly
| | - Mattia Chiesa
- Centro Cardiologico Monzino, IRCCsMilanItaly
- Department of Electronics, Information and Biomedical EngineeringPolitecnico di MilanoMilanItaly
| | | | - Michele Emdin
- Health Science Interdisciplinary Center, Scuola Superiore Sant'AnnaPisaItaly
- Cardio‐Thoracic DepartmentFondazione Toscana Gabriele MonasterioPisaItaly
| | - Massimo Piepoli
- Department of Clinical CardiologyIRCCS Policlinico San DonatoMilanItaly
- Department of Biomedical Sciences for HealthUniversity of MilanMilanItaly
| | - Gianfranco Sinagra
- Department of Cardiology‘Azienda Sanitaria Universitaria Giuliano‐Isontina’TriesteItaly
| | - Michele Senni
- Department of Cardiology, Unit of CardiologyASST Papa Giovanni XXIIIBergamoItaly
| | | | - Stamatis Adamopoulos
- Heart Failure and Heart Transplant UnitsOnassis Cardiac Surgery CentreAtticaGreece
| | - Dimitris Miliopoulos
- Heart Failure and Heart Transplant UnitsOnassis Cardiac Surgery CentreAtticaGreece
| | - Massimo Mapelli
- Centro Cardiologico Monzino, IRCCsMilanItaly
- Department of Clinical Sciences and Community Health, Section of CardiologyUniversity of MilanMilanItaly
| | | | | | | | | | | | - Damiano Magrì
- Department of Clinical and Molecular Medicine, Azienda Ospedaliera Sant'Andrea‘Sapienza’ Università degli Studi di RomaRomeItaly
| | - Stefania Paolillo
- Dipartimento di scienze biomediche avanzateFederico II UniversityNaplesItaly
| | - Ugo Corrà
- Department of Cardiology, Istituti Clinici Scientifici Maugeri, IRCCSVeruno InstituteVerunoItaly
| | - Rosa Raimondo
- Divisione di Cardiologia RiabilitativaIstituti Clinici Scientifici MaugeriVareseItaly
| | - Antonio Cittadini
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
- Interdepartmental Center for Gender Medicine Research ‘GENESIS’NaplesItaly
| | - Annamaria Iorio
- Department of Cardiology, Unit of CardiologyASST Papa Giovanni XXIIIBergamoItaly
| | - Andrea Salzano
- Cardiac UnitAORN A CardarelliNaplesItaly
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUK
| | - Rocco Lagioia
- UOC Cardiologia di RiabilitativaMater Dei HospitalBariItaly
| | | | - Roberto Badagliacca
- Dipartimento di Scienze Cliniche, Internistiche, Anestesiologiche e Cardiovascolari, ‘Sapienza’Rome UniversityRomeItaly
| | - Pasquale Perrone Filardi
- Department of Advanced Biomedical SciencesFederico II University of Naples and Mediterranea CardioCentroNaplesItaly
| | | | - Enrico Perna
- Dipartimento cardio‐toraco‐vascolareOspedale Cà Granda‐ A.O. NiguardaMilanItaly
| | - Marco Metra
- Department of Cardiology, Department of Medical and Surgical Specialities, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Gaia Cattadori
- Unità Operativa Cardiologia Riabilitativa, IRCCS MultimedicaMilanItaly
| | | | - Giuseppe Limongelli
- Cardiologia SUN, Ospedale Monaldi (Azienda dei Colli)Seconda Università di NapoliNaplesItaly
| | - Gianfranco Parati
- Department of Cardiovascular, Neural and Metabolic Sciences, San Luca HospitalIstituto Auxologico Italiano, IRCCSMilanItaly
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMilanItaly
| | | | - Maria Vittoria Matassini
- Department of Cardiology, Division of Cardiac Intensive Care Unit‐CardiologyOspedali Riuniti di AnconaAnconaItaly
| | - Francesco Bandera
- Department of Biomedical Sciences for HealthUniversity of MilanoMilanItaly
- Department of CardiologyIRCCS Policlinico San DonatoMilanItaly
| | - Maurizio Bussotti
- Cardiac Rehabilitation Unit, Istituti Clinici Scientifici Maugeri, IRCCSScientific Institute of MilanMilanItaly
| | - Federica Re
- Division of Cardiology, Cardiac Arrhythmia Center and Cardiomyopathies UnitSan Camillo‐Forlanini HospitalRomeItaly
| | - Carlo M. Lombardi
- Department of Cardiology, Department of Medical and Surgical Specialities, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | | | - Susanna Sciomer
- Dipartimento di Scienze Cliniche, Internistiche, Anestesiologiche e Cardiovascolari, ‘Sapienza’Rome UniversityRomeItaly
| | - Andrea Passantino
- Division of Cardiology, Istituti Clinici Scientifici MaugeriInstitute of BariBariItaly
| | | | - Davide Girola
- Clinica HildebrandCentro di Riabilitazione BrissagoBrissagoSwitzerland
| | - Claudio Passino
- Health Science Interdisciplinary Center, Scuola Superiore Sant'AnnaPisaItaly
| | - Marlus Karsten
- Centro Cardiologico Monzino, IRCCsMilanItaly
- Programa de Pós‐Graduação em Fisioterapia, UDESCFlorianópolisBrazil
| | - Savina Nodari
- Department of Medical and Surgical Specialities, Radiological Sciences and Public HealthUniversity of Brescia Medical SchoolBresciaItaly
| | - Giulio Pompilio
- Centro Cardiologico Monzino, IRCCsMilanItaly
- Department of Biomedical, Surgical and Dental SciencesUniversità degli Studi di MilanoMilanItaly
| | | |
Collapse
|
11
|
Sulaiman F, Khyriem C, Dsouza S, Abdul F, Alkhnbashi O, Faraji H, Farooqi M, Al Awadi F, Hassanein M, Ahmed F, Alsharhan M, Tawfik AR, Khamis AH, Bayoumi R. Characterizing Circulating microRNA Signatures of Type 2 Diabetes Subtypes. Int J Mol Sci 2025; 26:637. [PMID: 39859351 PMCID: PMC11766090 DOI: 10.3390/ijms26020637] [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: 11/21/2024] [Revised: 01/04/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Type 2 diabetes (T2D) is a heterogeneous disease influenced by both genetic and environmental factors. Recent studies suggest that T2D subtypes may exhibit distinct gene expression profiles. In this study, we aimed to identify T2D cluster-specific miRNA expression signatures for the previously reported five clinical subtypes that characterize the underlying pathophysiology of long-standing T2D: severe insulin-resistant diabetes (SIRD), severe insulin-deficient diabetes (SIDD), mild age-related diabetes (MARD), mild obesity-related diabetes (MOD), and mild early-onset diabetes (MEOD). We analyzed the circulating microRNAs (miRNAs) in 45 subjects representing the five T2D clusters and 7 non-T2D healthy controls by single-end small RNA sequencing. Bioinformatic analyses identified a total of 430 known circulating miRNAs and 13 previously unreported novel miRNAs. Of these, 71 were upregulated and 37 were downregulated in either controls or individual clusters. Each T2D subtype was associated with a specific dysregulated miRNA profile, distinct from that of healthy controls. Specifically, 3 upregulated miRNAs were unique to SIRD, 1 to MARD, 9 to MOD, and 18 to MEOD. Among the downregulated miRNAs, 11 were specific to SIRD, 9 to SIDD, 2 to MARD, and 1 to MEOD. Our study confirms the heterogeneity of T2D, represented by distinguishable subtypes both clinically and epigenetically and highlights the potential of miRNAs as markers for distinguishing the pathophysiology of T2D subtypes.
Collapse
Affiliation(s)
- Fatima Sulaiman
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| | - Costerwell Khyriem
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| | - Stafny Dsouza
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| | - Fatima Abdul
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| | - Omer Alkhnbashi
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates;
| | - Hanan Faraji
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| | - Muhammad Farooqi
- Dubai Diabetes Center, Dubai Health, Dubai P.O. Box 7272, United Arab Emirates;
| | - Fatheya Al Awadi
- Endocrinology Department, Dubai Hospital, Dubai Health, Dubai P.O. Box 7272, United Arab Emirates; (F.A.A.); (M.H.)
| | - Mohammed Hassanein
- Endocrinology Department, Dubai Hospital, Dubai Health, Dubai P.O. Box 7272, United Arab Emirates; (F.A.A.); (M.H.)
| | - Fayha Ahmed
- Pathology Department, Dubai Hospital, Dubai Health, Dubai P.O. Box 7272, United Arab Emirates; (F.A.); (M.A.)
| | - Mouza Alsharhan
- Pathology Department, Dubai Hospital, Dubai Health, Dubai P.O. Box 7272, United Arab Emirates; (F.A.); (M.A.)
| | - Abdel Rahman Tawfik
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (A.R.T.); (A.H.K.)
| | - Amar Hassan Khamis
- Hamdan Bin Mohammed College of Dental Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (A.R.T.); (A.H.K.)
| | - Riad Bayoumi
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates; (F.S.); (C.K.); (S.D.); (F.A.); (H.F.)
| |
Collapse
|
12
|
Wang J, Gao B, Wang J, Liu W, Yuan W, Chai Y, Ma J, Ma Y, Kong G, Liu M. Identifying subtypes of type 2 diabetes mellitus based on real-world electronic medical record data in China. Diabetes Res Clin Pract 2024; 217:111872. [PMID: 39332534 DOI: 10.1016/j.diabres.2024.111872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
AIMS To replicate the European subtypes of type 2 diabetes mellitus (T2DM) in the Chinese diabetes population and investigate the risk of complications in different subtypes. METHODS A diabetes cohort using real-world patient data was constructed, and clustering was employed to subgroup the T2DM patients. Kaplan-Meier analysis and the Cox models were used to analyze the association between diabetes subtypes and the risk of complications. RESULTS A total of 2,652 T2DM patients with complete clustering data were extracted. Among them, 466 (17.57 %) were classified as severe insulin-deficient diabetes (SIDD), 502 (18.93 %) as severe insulin-resistant diabetes (SIRD), 672 (25.34 %) as mild obesity-related diabetes (MOD), and 1,012 (38.16 %) as mild age-related diabetes (MARD). The risk of chronic kidney disease (CKD) and diabetic retinopathy (DR) were different in the four subtypes. Compared with MARD, SIRD had a higher risk of CKD (HR 2.40 [1.16, 4.96]), and SIDD had a higher risk of DR (HR 2.16 [1.11, 4.20]). The risk of stroke and coronary events had no difference. CONCLUSIONS The European T2DM subtypes can be replicated in the Chinese diabetes population. The risk of CKD and DR varied among different subtypes, indicating that proper interventions can be taken to prevent specific complications in different subtypes.
Collapse
Affiliation(s)
- Jiayu Wang
- National Institute of Health Data Science, Peking University, Beijing 100191, China; Advanced Institute of Information Technology, Peking University, Hangzhou 314201, Zhejiang, China; Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China
| | - Bixia Gao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Jinwei Wang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Wenwen Liu
- National Institute of Health Data Science, Peking University, Beijing 100191, China
| | - Weijia Yuan
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China
| | - Yangfan Chai
- Peking University Chongqing Research Institute of Big Data, Chongqing 100871, China
| | - Jun Ma
- National Institute of Health Data Science, Peking University, Beijing 100191, China
| | - Yangyang Ma
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China
| | - Guilan Kong
- National Institute of Health Data Science, Peking University, Beijing 100191, China; Advanced Institute of Information Technology, Peking University, Hangzhou 314201, Zhejiang, China; Institute of Medical Technology, Peking University Health Science Center, Beijing 100191, China.
| | - Minchao Liu
- Department of Computer Application and Management, Chinese PLA General Hospital, Beijing 100039, China.
| |
Collapse
|
13
|
Navratilova HF, Whetton AD, Geifman N. Artificial intelligence driven definition of food preference endotypes in UK Biobank volunteers is associated with distinctive health outcomes and blood based metabolomic and proteomic profiles. J Transl Med 2024; 22:881. [PMID: 39354608 PMCID: PMC11443809 DOI: 10.1186/s12967-024-05663-0] [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: 05/29/2024] [Accepted: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Specific food preferences can determine an individual's dietary patterns and therefore, may be associated with certain health risks and benefits. METHODS Using food preference questionnaire (FPQ) data from a subset comprising over 180,000 UK Biobank participants, we employed Latent Profile Analysis (LPA) approach to identify the main patterns or profiles among participants. blood biochemistry across groups/profiles was compared using the non-parametric Kruskal-Wallis test. We applied the Limma algorithm for differential abundance analysis on 168 metabolites and 2923 proteins, and utilized the Database for Annotation, Visualization and Integrated Discovery (DAVID) to identify enriched biological processes and pathways. Relative risks (RR) were calculated for chronic diseases and mental conditions per group, adjusting for sociodemographic factors. RESULTS Based on their food preferences, three profiles were termed: the putative Health-conscious group (low preference for animal-based or sweet foods, and high preference for vegetables and fruits), the Omnivore group (high preference for all foods), and the putative Sweet-tooth group (high preference for sweet foods and sweetened beverages). The Health-conscious group exhibited lower risk of heart failure (RR = 0.86, 95%CI 0.79-0.93) and chronic kidney disease (RR = 0.69, 95%CI 0.65-0.74) compared to the two other groups. The Sweet-tooth group had greater risk of depression (RR = 1.27, 95%CI 1.21-1.34), diabetes (RR = 1.15, 95%CI 1.01-1.31), and stroke (RR = 1.22, 95%CI 1.15-1.31) compared to the other two groups. Cancer (overall) relative risk showed little difference across the Health-conscious, Omnivore, and Sweet-tooth groups with RR of 0.98 (95%CI 0.96-1.01), 1.00 (95%CI 0.98-1.03), and 1.01 (95%CI 0.98-1.04), respectively. The Health-conscious group was associated with lower levels of inflammatory biomarkers (e.g., C-reactive Protein) which are also known to be elevated in those with common metabolic diseases (e.g., cardiovascular disease). Other markers modulated in the Health-conscious group, ketone bodies, insulin-like growth factor-binding protein (IGFBP), and Growth Hormone 1 were more abundant, while leptin was less abundant. Further, the IGFBP pathway, which influences IGF1 activity, may be significantly enhanced by dietary choices. CONCLUSIONS These observations align with previous findings from studies focusing on weight loss interventions, which include a reduction in leptin levels. Overall, the Health-conscious group, with preference to healthier food options, has better health outcomes, compared to Sweet-tooth and Omnivore groups.
Collapse
Affiliation(s)
- Hana F Navratilova
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
- Veterinary Health Innovation Engine, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, UK
- Department of Community Nutrition, Faculty of Human Ecology, IPB University, Bogor, 16680, Indonesia
| | - Anthony D Whetton
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
- Veterinary Health Innovation Engine, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, UK
| | - Nophar Geifman
- Veterinary Health Innovation Engine, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, UK
- School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7YH, UK
| |
Collapse
|
14
|
Xian S, Grabowska ME, Kullo IJ, Luo Y, Smoller JW, Wei WQ, Jarvik G, Mooney S, Crosslin D. Language-model-based patient embedding using electronic health records facilitates phenotyping, disease forecasting, and progression analysis. RESEARCH SQUARE 2024:rs.3.rs-4708839. [PMID: 39399661 PMCID: PMC11469380 DOI: 10.21203/rs.3.rs-4708839/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Current studies regarding the secondary use of electronic health records (EHR) predominantly rely on domain expertise and existing medical knowledge. Though significant efforts have been devoted to investigating the application of machine learning algorithms in the EHR, efficient and powerful representation of patients is needed to unleash the potential of discovering new medical patterns underlying the EHR. Here, we present an unsupervised method for embedding high-dimensional EHR data at the patient level, aimed at characterizing patient heterogeneity in complex diseases and identifying new disease patterns associated with clinical outcome disparities. Inspired by the architecture of modern language models-specifically transformers with attention mechanisms, we use patient diagnosis and procedure codes as vocabularies and treat each patient as a sentence to perform the patient embedding. We applied this approach to 34,851 unique medical codes across 1,046,649 longitudinal patient events, including 102,739 patients from the electronic Medical Records and GEnomics (eMERGE) Network. The resulting patient vectors demonstrated excellent performance in predicting future disease events (median AUROC = 0.87 within one year) and bulk phenotyping (median AUROC = 0.84). We then illustrated the utility of these patient vectors in revealing heterogeneous comorbidity patterns, exemplified by disease subtypes in colorectal cancer and systemic lupus erythematosus, and capturing distinct longitudinal disease trajectories. External validation using EHR data from the University of Washington confirmed robust model performance, with median AUROCs of 0.83 and 0.84 for bulk phenotyping tasks and disease onset prediction, respectively. Importantly, the model reproduced the clustering results of disease subtypes identified in the eMERGE cohort and uncovered variations in overall mortality among these subtypes. Together, these results underscore the potential of representation learning in EHRs to enhance patient characterization and associated clinical outcomes, thereby advancing disease forecasting and facilitating personalized medicine.
Collapse
Affiliation(s)
- Su Xian
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA
| | - Monika E Grabowska
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine and the Gonda Vascular Center, Mayo Clinic Rochester Minnesota
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Gail Jarvik
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA
| | - Sean Mooney
- Center for Information Technology, National Institutes of Health
| | - David Crosslin
- Department of Medicine, Division of Biomedical Informatics and Genomics, Tulane University, New Orleans, LA
| |
Collapse
|
15
|
Ryan B, Marioni RE, Simpson TI. Multi-Omic Graph Diagnosis (MOGDx): a data integration tool to perform classification tasks for heterogeneous diseases. BIOINFORMATICS (OXFORD, ENGLAND) 2024; 40:btae523. [PMID: 39177104 PMCID: PMC11374023 DOI: 10.1093/bioinformatics/btae523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/17/2024] [Accepted: 08/23/2024] [Indexed: 08/24/2024]
Abstract
MOTIVATION Heterogeneity in human diseases presents challenges in diagnosis and treatments due to the broad range of manifestations and symptoms. With the rapid development of labelled multi-omic data, integrative machine learning methods have achieved breakthroughs in treatments by redefining these diseases at a more granular level. These approaches often have limitations in scalability, oversimplification, and handling of missing data. RESULTS In this study, we introduce Multi-Omic Graph Diagnosis (MOGDx), a flexible command line tool for the integration of multi-omic data to perform classification tasks for heterogeneous diseases. MOGDx has a network taxonomy. It fuses patient similarity networks, augments this integrated network with a reduced vector representation of genomic data and performs classification using a graph convolutional network. MOGDx was evaluated on three datasets from the cancer genome atlas for breast invasive carcinoma, kidney cancer, and low grade glioma. MOGDx demonstrated state-of-the-art performance and an ability to identify relevant multi-omic markers in each task. It integrated more genomic measures with greater patient coverage compared to other network integrative methods. Overall, MOGDx is a promising tool for integrating multi-omic data, classifying heterogeneous diseases, and aiding interpretation of genomic marker data. AVAILABILITY AND IMPLEMENTATION MOGDx source code is available from https://github.com/biomedicalinformaticsgroup/MOGDx.
Collapse
Affiliation(s)
- Barry Ryan
- School of Informatics, University of Edinburgh, 10 Crichton Street, Edinburgh, EH8 9AB, United Kingdom
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - T Ian Simpson
- School of Informatics, University of Edinburgh, 10 Crichton Street, Edinburgh, EH8 9AB, United Kingdom
| |
Collapse
|
16
|
Sirota M, Kodama L, Woldemariam S, Tang A, Li Y, Kornak J, Allen IE, Raphael E, Oskotsky T. Sex-stratified analyses of comorbidities associated with an inpatient delirium diagnosis using real world data. RESEARCH SQUARE 2024:rs.3.rs-4765249. [PMID: 39108477 PMCID: PMC11302686 DOI: 10.21203/rs.3.rs-4765249/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Delirium is a detrimental mental condition often seen in older, hospitalized patients and is currently hard to predict. In this study, we leverage electronic health records (EHR) to identify 7,492 UCSF patients and 19,417 UC health system patients with an inpatient delirium diagnosis and the same number of control patients without delirium. We found significant associations between comorbidities or laboratory values and an inpatient delirium diagnosis, including metabolic abnormalities and psychiatric diagnoses. Some associations were sex-specific, including dementia subtypes and infections. We further explored the associations with anemia and bipolar disorder by conducting longitudinal analyses from the time of first diagnosis to development of delirium, demonstrating a significant relationship across time. Finally, we show that an inpatient delirium diagnosis leads to increased risk of mortality. These results demonstrate the powerful application of the EHR to shed insights into prior diagnoses and laboratory values that could help predict development of inpatient delirium and the importance of sex when making these assessments.
Collapse
|
17
|
Su C, Hou Y, Xu J, Xu Z, Zhou M, Ke A, Li H, Xu J, Brendel M, Maasch JRMA, Bai Z, Zhang H, Zhu Y, Cincotta MC, Shi X, Henchcliffe C, Leverenz JB, Cummings J, Okun MS, Bian J, Cheng F, Wang F. Identification of Parkinson's disease PACE subtypes and repurposing treatments through integrative analyses of multimodal data. NPJ Digit Med 2024; 7:184. [PMID: 38982243 PMCID: PMC11233682 DOI: 10.1038/s41746-024-01175-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 06/21/2024] [Indexed: 07/11/2024] Open
Abstract
Parkinson's disease (PD) is a serious neurodegenerative disorder marked by significant clinical and progression heterogeneity. This study aimed at addressing heterogeneity of PD through integrative analysis of various data modalities. We analyzed clinical progression data (≥5 years) of individuals with de novo PD using machine learning and deep learning, to characterize individuals' phenotypic progression trajectories for PD subtyping. We discovered three pace subtypes of PD exhibiting distinct progression patterns: the Inching Pace subtype (PD-I) with mild baseline severity and mild progression speed; the Moderate Pace subtype (PD-M) with mild baseline severity but advancing at a moderate progression rate; and the Rapid Pace subtype (PD-R) with the most rapid symptom progression rate. We found cerebrospinal fluid P-tau/α-synuclein ratio and atrophy in certain brain regions as potential markers of these subtypes. Analyses of genetic and transcriptomic profiles with network-based approaches identified molecular modules associated with each subtype. For instance, the PD-R-specific module suggested STAT3, FYN, BECN1, APOA1, NEDD4, and GATA2 as potential driver genes of PD-R. It also suggested neuroinflammation, oxidative stress, metabolism, PI3K/AKT, and angiogenesis pathways as potential drivers for rapid PD progression (i.e., PD-R). Moreover, we identified repurposable drug candidates by targeting these subtype-specific molecular modules using network-based approach and cell line drug-gene signature data. We further estimated their treatment effects using two large-scale real-world patient databases; the real-world evidence we gained highlighted the potential of metformin in ameliorating PD progression. In conclusion, this work helps better understand clinical and pathophysiological complexity of PD progression and accelerate precision medicine.
Collapse
Grants
- R21 AG083003 NIA NIH HHS
- R01 AG082118 NIA NIH HHS
- R56 AG074001 NIA NIH HHS
- R01AG076448 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1AG072449 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- MJFF-023081 Michael J. Fox Foundation for Parkinson's Research (Michael J. Fox Foundation)
- R01AG080991 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P30 AG072959 NIA NIH HHS
- 3R01AG066707-01S1 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R21AG083003 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01AG066707 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R35 AG071476 NIA NIH HHS
- RF1 AG082211 NIA NIH HHS
- R56AG074001 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01AG082118 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R25 AG083721 NIA NIH HHS
- RF1AG082211 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 NS093334 NINDS NIH HHS
- AG083721-01 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1NS133812 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P20GM109025 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1 NS133812 NINDS NIH HHS
- R35AG71476 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01 AG073323 NIA NIH HHS
- R01 AG066707 NIA NIH HHS
- R01AG053798 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01AG076234 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01 AG076448 NIA NIH HHS
- R01 AG080991 NIA NIH HHS
- R01 AG076234 NIA NIH HHS
- U01NS093334 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- P20 GM109025 NIGMS NIH HHS
- P30AG072959 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- RF1 AG072449 NIA NIH HHS
- R01 AG053798 NIA NIH HHS
- 3R01AG066707-02S1 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- U01AG073323 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- ALZDISCOVERY-1051936 Alzheimer's Association
Collapse
Affiliation(s)
- Chang Su
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Yu Hou
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Jielin Xu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Zhenxing Xu
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Manqi Zhou
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Alison Ke
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Haoyang Li
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jie Xu
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Matthew Brendel
- Institute for Computational Biomedicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jacqueline R M A Maasch
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Computer Science, Cornell Tech, Cornell University, New York, NY, USA
| | - Zilong Bai
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Haotan Zhang
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Yingying Zhu
- Department of Computer Science, University of Texas at Arlington, Arlington, TX, USA
| | - Molly C Cincotta
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xinghua Shi
- Department of Computer and Information Sciences, Temple University, Philadelphia, PA, USA
| | - Claire Henchcliffe
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Michael S Okun
- Department of Neurology, Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Institute of Artificial Intelligence for Digital Health, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
18
|
Tang AS, Woldemariam SR, Miramontes S, Norgeot B, Oskotsky TT, Sirota M. Harnessing EHR data for health research. Nat Med 2024; 30:1847-1855. [PMID: 38965433 DOI: 10.1038/s41591-024-03074-8] [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: 01/03/2024] [Accepted: 05/17/2024] [Indexed: 07/06/2024]
Abstract
With the increasing availability of rich, longitudinal, real-world clinical data recorded in electronic health records (EHRs) for millions of patients, there is a growing interest in leveraging these records to improve the understanding of human health and disease and translate these insights into clinical applications. However, there is also a need to consider the limitations of these data due to various biases and to understand the impact of missing information. Recognizing and addressing these limitations can inform the design and interpretation of EHR-based informatics studies that avoid confusing or incorrect conclusions, particularly when applied to population or precision medicine. Here we discuss key considerations in the design, implementation and interpretation of EHR-based informatics studies, drawing from examples in the literature across hypothesis generation, hypothesis testing and machine learning applications. We outline the growing opportunities for EHR-based informatics studies, including association studies and predictive modeling, enabled by evolving AI capabilities-while addressing limitations and potential pitfalls to avoid.
Collapse
Affiliation(s)
- Alice S Tang
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Sarah R Woldemariam
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Silvia Miramontes
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | | | - Tomiko T Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
19
|
Ojima T, Namba S, Suzuki K, Yamamoto K, Sonehara K, Narita A, Kamatani Y, Tamiya G, Yamamoto M, Yamauchi T, Kadowaki T, Okada Y. Body mass index stratification optimizes polygenic prediction of type 2 diabetes in cross-biobank analyses. Nat Genet 2024; 56:1100-1109. [PMID: 38862855 DOI: 10.1038/s41588-024-01782-y] [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: 07/17/2022] [Accepted: 04/26/2024] [Indexed: 06/13/2024]
Abstract
Type 2 diabetes (T2D) shows heterogeneous body mass index (BMI) sensitivity. Here, we performed stratification based on BMI to optimize predictions for BMI-related diseases. We obtained BMI-stratified datasets using data from more than 195,000 individuals (nT2D = 55,284) from BioBank Japan (BBJ) and UK Biobank. T2D heritability in the low-BMI group was greater than that in the high-BMI group. Polygenic predictions of T2D toward low-BMI targets had pseudo-R2 values that were more than 22% higher than BMI-unstratified targets. Polygenic risk scores (PRSs) from low-BMI discovery outperformed PRSs from high BMI, while PRSs from BMI-unstratified discovery performed best. Pathway-specific PRSs demonstrated the biological contributions of pathogenic pathways. Low-BMI T2D cases showed higher rates of neuropathy and retinopathy. Combining BMI stratification and a method integrating cross-population effects, T2D predictions showed greater than 37% improvements over unstratified-matched-population prediction. We replicated findings in the Tohoku Medical Megabank (n = 26,000) and the second BBJ cohort (n = 33,096). Our findings suggest that target stratification based on existing traits can improve the polygenic prediction of heterogeneous diseases.
Collapse
Affiliation(s)
- Takafumi Ojima
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Center for Advanced Intelligence Project, RIKEN, Tokyo, Japan
| | - Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ken Suzuki
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Laboratory of Children's Health and Genetics, Division of Health Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kyuto Sonehara
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akira Narita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Yoichiro Kamatani
- Laboratory of Complex Trait Genomics, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Gen Tamiya
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Center for Advanced Intelligence Project, RIKEN, Tokyo, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Masayuki Yamamoto
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan.
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka, Japan.
| |
Collapse
|
20
|
Wu X, Luo G, Dong Z, Zheng W, Jia G. Integrated Pleiotropic Gene Set Unveils Comorbidity Insights across Digestive Cancers and Other Diseases. Genes (Basel) 2024; 15:478. [PMID: 38674412 PMCID: PMC11049963 DOI: 10.3390/genes15040478] [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: 03/09/2024] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Comorbidities are prevalent in digestive cancers, intensifying patient discomfort and complicating prognosis. Identifying potential comorbidities and investigating their genetic connections in a systemic manner prove to be instrumental in averting additional health challenges during digestive cancer management. Here, we investigated 150 diseases across 18 categories by collecting and integrating various factors related to disease comorbidity, such as disease-associated SNPs or genes from sources like MalaCards, GWAS Catalog and UK Biobank. Through this extensive analysis, we have established an integrated pleiotropic gene set comprising 548 genes in total. Particularly, there enclosed the genes encoding major histocompatibility complex or related to antigen presentation. Additionally, we have unveiled patterns in protein-protein interactions and key hub genes/proteins including TP53, KRAS, CTNNB1 and PIK3CA, which may elucidate the co-occurrence of digestive cancers with certain diseases. These findings provide valuable insights into the molecular origins of comorbidity, offering potential avenues for patient stratification and the development of targeted therapies in clinical trials.
Collapse
Affiliation(s)
- Xinnan Wu
- Institute of Public-Safety and Big Data, College of Data Science, Taiyuan University of Technology, University Street, Yuci District, Jinzhong 030600, China;
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; (G.L.); (Z.D.)
| | - Guangwen Luo
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; (G.L.); (Z.D.)
| | - Zhaonian Dong
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; (G.L.); (Z.D.)
| | - Wen Zheng
- Institute of Public-Safety and Big Data, College of Data Science, Taiyuan University of Technology, University Street, Yuci District, Jinzhong 030600, China;
| | - Gengjie Jia
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; (G.L.); (Z.D.)
| |
Collapse
|
21
|
Sahu M, Vashishth S, Kukreti N, Gulia A, Russell A, Ambasta RK, Kumar P. Synergizing drug repurposing and target identification for neurodegenerative diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 205:111-169. [PMID: 38789177 DOI: 10.1016/bs.pmbts.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Despite dedicated research efforts, the absence of disease-curing remedies for neurodegenerative diseases (NDDs) continues to jeopardize human society and stands as a challenge. Drug repurposing is an attempt to find new functionality of existing drugs and take it as an opportunity to discourse the clinically unmet need to treat neurodegeneration. However, despite applying this approach to rediscover a drug, it can also be used to identify the target on which a drug could work. The primary objective of target identification is to unravel all the possibilities of detecting a new drug or repurposing an existing drug. Lately, scientists and researchers have been focusing on specific genes, a particular site in DNA, a protein, or a molecule that might be involved in the pathogenesis of the disease. However, the new era discusses directing the signaling mechanism involved in the disease progression, where receptors, ion channels, enzymes, and other carrier molecules play a huge role. This review aims to highlight how target identification can expedite the whole process of drug repurposing. Here, we first spot various target-identification methods and drug-repositioning studies, including drug-target and structure-based identification studies. Moreover, we emphasize various drug repurposing approaches in NDDs, namely, experimental-based, mechanism-based, and in silico approaches. Later, we draw attention to validation techniques and stress on drugs that are currently undergoing clinical trials in NDDs. Lastly, we underscore the future perspective of synergizing drug repurposing and target identification in NDDs and present an unresolved question to address the issue.
Collapse
Affiliation(s)
- Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Shrutikirti Vashishth
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Neha Kukreti
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Ashima Gulia
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Ashish Russell
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Rashmi K Ambasta
- Department of Biotechnology and Microbiology, SRM University, Sonepat, Haryana, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, Delhi, India.
| |
Collapse
|
22
|
Mizuno S, Wagata M, Nagaie S, Ishikuro M, Obara T, Tamiya G, Kuriyama S, Tanaka H, Yaegashi N, Yamamoto M, Sugawara J, Ogishima S. Development of phenotyping algorithms for hypertensive disorders of pregnancy (HDP) and their application in more than 22,000 pregnant women. Sci Rep 2024; 14:6292. [PMID: 38491024 PMCID: PMC10943000 DOI: 10.1038/s41598-024-55914-9] [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: 06/15/2023] [Accepted: 02/28/2024] [Indexed: 03/18/2024] Open
Abstract
Recently, many phenotyping algorithms for high-throughput cohort identification have been developed. Prospective genome cohort studies are critical resources for precision medicine, but there are many hurdles in the precise cohort identification. Consequently, it is important to develop phenotyping algorithms for cohort data collection. Hypertensive disorders of pregnancy (HDP) is a leading cause of maternal morbidity and mortality. In this study, we developed, applied, and validated rule-based phenotyping algorithms of HDP. Two phenotyping algorithms, algorithms 1 and 2, were developed according to American and Japanese guidelines, and applied into 22,452 pregnant women in the Birth and Three-Generation Cohort Study of the Tohoku Medical Megabank project. To precise cohort identification, we analyzed both structured data (e.g., laboratory and physiological tests) and unstructured clinical notes. The identified subtypes of HDP were validated against reference standards. Algorithms 1 and 2 identified 7.93% and 8.08% of the subjects as having HDP, respectively, along with their HDP subtypes. Our algorithms were high performing with high positive predictive values (0.96 and 0.90 for algorithms 1 and 2, respectively). Overcoming the hurdle of precise cohort identification from large-scale cohort data collection, we achieved both developed and implemented phenotyping algorithms, and precisely identified HDP patients and their subtypes from large-scale cohort data collection.
Collapse
Affiliation(s)
- Satoshi Mizuno
- Department of Informatics for Genomic Medicine, Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Maiko Wagata
- Department of Feto-Maternal Medical Science, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
| | - Satoshi Nagaie
- Department of Informatics for Genomic Medicine, Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Mami Ishikuro
- Department of Molecular Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
| | - Taku Obara
- Department of Molecular Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
| | - Gen Tamiya
- Department of Statistical Genetics and Genomics, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
| | - Shinichi Kuriyama
- Department of Molecular Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
| | | | - Nobuo Yaegashi
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Tohoku University, Miyagi, Japan
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
| | - Junichi Sugawara
- Department of Gynecology and Obstetrics, Tohoku University Graduate School of Medicine, Tohoku University, Miyagi, Japan
- Suzuki Memorial Hospital, 3-5-5, Satonomori, Iwanumashi, Miyagi, Japan
| | - Soichi Ogishima
- Department of Informatics for Genomic Medicine, Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan.
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Miyagi, Japan.
| |
Collapse
|
23
|
Chafai N, Bonizzi L, Botti S, Badaoui B. Emerging applications of machine learning in genomic medicine and healthcare. Crit Rev Clin Lab Sci 2024; 61:140-163. [PMID: 37815417 DOI: 10.1080/10408363.2023.2259466] [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: 06/19/2023] [Accepted: 09/12/2023] [Indexed: 10/11/2023]
Abstract
The integration of artificial intelligence technologies has propelled the progress of clinical and genomic medicine in recent years. The significant increase in computing power has facilitated the ability of artificial intelligence models to analyze and extract features from extensive medical data and images, thereby contributing to the advancement of intelligent diagnostic tools. Artificial intelligence (AI) models have been utilized in the field of personalized medicine to integrate clinical data and genomic information of patients. This integration allows for the identification of customized treatment recommendations, ultimately leading to enhanced patient outcomes. Notwithstanding the notable advancements, the application of artificial intelligence (AI) in the field of medicine is impeded by various obstacles such as the limited availability of clinical and genomic data, the diversity of datasets, ethical implications, and the inconclusive interpretation of AI models' results. In this review, a comprehensive evaluation of multiple machine learning algorithms utilized in the fields of clinical and genomic medicine is conducted. Furthermore, we present an overview of the implementation of artificial intelligence (AI) in the fields of clinical medicine, drug discovery, and genomic medicine. Finally, a number of constraints pertaining to the implementation of artificial intelligence within the healthcare industry are examined.
Collapse
Affiliation(s)
- Narjice Chafai
- Laboratory of Biodiversity, Ecology, and Genome, Faculty of Sciences, Department of Biology, Mohammed V University in Rabat, Rabat, Morocco
| | - Luigi Bonizzi
- Department of Biomedical, Surgical and Dental Science, University of Milan, Milan, Italy
| | - Sara Botti
- PTP Science Park, Via Einstein - Loc. Cascina Codazza, Lodi, Italy
| | - Bouabid Badaoui
- Laboratory of Biodiversity, Ecology, and Genome, Faculty of Sciences, Department of Biology, Mohammed V University in Rabat, Rabat, Morocco
- African Sustainable Agriculture Research Institute (ASARI), Mohammed VI Polytechnic University (UM6P), Laâyoune, Morocco
| |
Collapse
|
24
|
Myasoedova VA, Chiesa M, Cosentino N, Bonomi A, Ludergnani M, Bozzi M, Valerio V, Moschetta D, Massaiu I, Mantegazza V, Marenzi G, Poggio P. Non-stenotic fibro-calcific aortic valve as a predictor of myocardial infarction recurrence. Eur J Prev Cardiol 2024:zwae062. [PMID: 38365224 DOI: 10.1093/eurjpc/zwae062] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/18/2024]
Abstract
BACKGROUND Patients with acute myocardial infarction (AMI) are at increased risk of recurrent cardiovascular events. Non-stenotic aortic valve fibro-calcific remodeling (AVSc), reflecting systemic damage, may serve as a new marker of risk. OBJECTIVES To stratify subgroups of AMI patients with specific probabilities of recurrent AMI and to evaluate the importance of AVSc in this setting. METHODS Consecutive AMI patients (n = 2530) were admitted at Centro Cardiologico Monzino (2010-2019) and followed up for 5 years. Patients were divided into study (n = 1070) and test (n = 966) cohorts. Topological data analysis (TDA) was used to stratify patient subgroups, while Kaplan-Meier and Cox regressions analyses were used to evaluate the significance of baseline characteristics. RESULTS TDA identified 11 subgroups of AMI patients with specific baseline characteristics. Two subgroups showed the highest rate of reinfarction after 5 years from the indexed AMI with a combined hazard ratio (HR) of 3.8 (95%CI: 2.7-5.4) compared to the other subgroups. This was confirmed in the test cohort (HR = 3.1; 95%CI: 2.2-4.3). These two subgroups were mostly men, with hypertension and dyslipidemia, who exhibit higher prevalence of AVSc, higher levels of high-sensitive c-reactive protein and creatinine. In the year-by-year analysis, AVSc, adjusted for all confounders, showed an independent association with the increased risk of reinfarction (odds ratio of ∼2 at all time-points), in both the study and the test cohorts (all p < 0.01). CONCLUSIONS AVSc is a crucial variable for identifying AMI patients at high risk of recurrent AMI and its presence should be considered when assessing the management of AMI patients. The inclusion of AVSc in risk stratification models may improve the accuracy of predicting the likelihood of recurrent AMI, leading to more personalized treatment decisions.
Collapse
Affiliation(s)
| | - Mattia Chiesa
- Centro Cardiologico Monzino, IRCCS, Milan Italy
- Department of Electronics, Information and Biomedical engineering, Politecnico di Milano, Milan, Italy
| | - Nicola Cosentino
- Centro Cardiologico Monzino, IRCCS, Milan Italy
- Department of Clinical Sciences and Community Health, Cardiovascular Section, University of Milan, Milan, Italy
| | | | | | | | | | | | | | - Valentina Mantegazza
- Centro Cardiologico Monzino, IRCCS, Milan Italy
- Department of Clinical Sciences and Community Health, Cardiovascular Section, University of Milan, Milan, Italy
| | | | - Paolo Poggio
- Centro Cardiologico Monzino, IRCCS, Milan Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milano, Italy
| |
Collapse
|
25
|
Nymand L, Nielsen ML, Vittrup I, Halling AS, Francis Thomsen S, Egeberg A, Thyssen JP. Atopic dermatitis phenotypes based on cluster analysis of the Danish Skin Cohort. Br J Dermatol 2024; 190:207-215. [PMID: 37850907 DOI: 10.1093/bjd/ljad401] [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: 05/04/2023] [Revised: 10/04/2023] [Accepted: 10/14/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Despite previous attempts to classify atopic dermatitis (AD) into subtypes (e.g. extrinsic vs. intrinsic), there is a need to better understand specific phenotypes in adulthood. OBJECTIVES To identify, using machine learning (ML), adult AD phenotypes. METHODS We used unsupervised cluster analysis to identify AD phenotypes by analysing different responses to predetermined variables (age of disease onset, severity, itch and skin pain intensity, flare frequency, anatomical location, presence and/or severity of current comorbidities) in adults with AD from the Danish Skin Cohort. RESULTS The unsupervised cluster analysis resulted in five clusters where AD severity most clearly differed. We classified them as 'mild', 'mild-to-moderate', 'moderate', 'severe' and 'very severe'. The severity of multiple predetermined patient-reported outcomes was positively associated with AD, including an increased number of flare-ups and increased flare-up duration and disease severity. However, an increased severity of rhinitis and mental health burden was also found for the mild-to-moderate phenotype. CONCLUSIONS ML confirmed the use of disease severity for the categorization of phenotypes, and our cluster analysis provided novel detailed information about how flare patterns and duration are associated with AD disease severity.
Collapse
Affiliation(s)
- Lea Nymand
- Department of Dermatology, Bispebjerg Hospital
| | | | - Ida Vittrup
- Department of Dermatology, Bispebjerg Hospital
| | | | - Simon Francis Thomsen
- Department of Dermatology, Bispebjerg Hospital
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences
| | - Alexander Egeberg
- Department of Dermatology, Bispebjerg Hospital
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jacob P Thyssen
- Department of Dermatology, Bispebjerg Hospital
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Li H, Zhou M, Sun Y, Yang J, Zeng X, Qiu Y, Xia Y, Zheng Z, Yu J, Feng Y, Shi Z, Huang T, Tan L, Lin R, Li J, Fan X, Ye J, Duan H, Shi S, Shu Q. A Patient Similarity Network (CHDmap) to Predict Outcomes After Congenital Heart Surgery: Development and Validation Study. JMIR Med Inform 2024; 12:e49138. [PMID: 38297829 PMCID: PMC10850852 DOI: 10.2196/49138] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/21/2023] [Accepted: 11/16/2023] [Indexed: 02/02/2024] Open
Abstract
Background Although evidence-based medicine proposes personalized care that considers the best evidence, it still fails to address personal treatment in many real clinical scenarios where the complexity of the situation makes none of the available evidence applicable. "Medicine-based evidence" (MBE), in which big data and machine learning techniques are embraced to derive treatment responses from appropriately matched patients in real-world clinical practice, was proposed. However, many challenges remain in translating this conceptual framework into practice. Objective This study aimed to technically translate the MBE conceptual framework into practice and evaluate its performance in providing general decision support services for outcomes after congenital heart disease (CHD) surgery. Methods Data from 4774 CHD surgeries were collected. A total of 66 indicators and all diagnoses were extracted from each echocardiographic report using natural language processing technology. Combined with some basic clinical and surgical information, the distances between each patient were measured by a series of calculation formulas. Inspired by structure-mapping theory, the fusion of distances between different dimensions can be modulated by clinical experts. In addition to supporting direct analogical reasoning, a machine learning model can be constructed based on similar patients to provide personalized prediction. A user-operable patient similarity network (PSN) of CHD called CHDmap was proposed and developed to provide general decision support services based on the MBE approach. Results Using 256 CHD cases, CHDmap was evaluated on 2 different types of postoperative prognostic prediction tasks: a binary classification task to predict postoperative complications and a multiple classification task to predict mechanical ventilation duration. A simple poll of the k-most similar patients provided by the PSN can achieve better prediction results than the average performance of 3 clinicians. Constructing logistic regression models for prediction using similar patients obtained from the PSN can further improve the performance of the 2 tasks (best area under the receiver operating characteristic curve=0.810 and 0.926, respectively). With the support of CHDmap, clinicians substantially improved their predictive capabilities. Conclusions Without individual optimization, CHDmap demonstrates competitive performance compared to clinical experts. In addition, CHDmap has the advantage of enabling clinicians to use their superior cognitive abilities in conjunction with it to make decisions that are sometimes even superior to those made using artificial intelligence models. The MBE approach can be embraced in clinical practice, and its full potential can be realized.
Collapse
Affiliation(s)
- Haomin Li
- Clinical Data Center, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Mengying Zhou
- Clinical Data Center, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Yuhan Sun
- Clinical Data Center, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Jian Yang
- Clinical Data Center, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Xian Zeng
- Clinical Data Center, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Yunxiang Qiu
- Cardiac Intensive Care Unit, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuanyuan Xia
- Cardiac Intensive Care Unit, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Zhijie Zheng
- Cardiac Intensive Care Unit, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jin Yu
- Ultrasonography Department, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuqing Feng
- Clinical Data Center, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Zhuo Shi
- Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ting Huang
- Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Linhua Tan
- Cardiac Intensive Care Unit, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ru Lin
- Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianhua Li
- Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiangming Fan
- Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jingjing Ye
- Ultrasonography Department, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Huilong Duan
- The College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Shanshan Shi
- Cardiac Intensive Care Unit, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qiang Shu
- Cardiac Surgery, The Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
27
|
Sajedi S, Ebrahimi G, Roudi R, Mehta I, Heshmat A, Samimi H, Kazempour S, Zainulabadeen A, Docking TR, Arora SP, Cigarroa F, Seshadri S, Karsan A, Zare H. Integrating DNA methylation and gene expression data in a single gene network using the iNETgrate package. Sci Rep 2023; 13:21721. [PMID: 38066050 PMCID: PMC10709411 DOI: 10.1038/s41598-023-48237-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Analyzing different omics data types independently is often too restrictive to allow for detection of subtle, but consistent, variations that are coherently supported based upon different assays. Integrating multi-omics data in one model can increase statistical power. However, designing such a model is challenging because different omics are measured at different levels. We developed the iNETgrate package ( https://bioconductor.org/packages/iNETgrate/ ) that efficiently integrates transcriptome and DNA methylation data in a single gene network. Applying iNETgrate on five independent datasets improved prognostication compared to common clinical gold standards and a patient similarity network approach.
Collapse
Affiliation(s)
- Sogand Sajedi
- Department of Cell Systems & Anatomy, The University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, San Antonio, TX, 78229, USA
| | - Ghazal Ebrahimi
- Bioinformatics Program, The University of British Columbia, Vancouver, BC, Canada
| | - Raheleh Roudi
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Isha Mehta
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Amirreza Heshmat
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hanie Samimi
- School of Architecture, University of Utah, Salt Lake City, UT, 84112, USA
| | - Shiva Kazempour
- Department of Cell Systems & Anatomy, The University of Texas Health Science Center, San Antonio, TX, 78229, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, San Antonio, TX, 78229, USA
| | - Aamir Zainulabadeen
- Department of Computer Science, Princeton University, Princeton, NJ, 08540, USA
| | - Thomas Roderick Docking
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada
| | - Sukeshi Patel Arora
- Mays Cancer Center, The University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Francisco Cigarroa
- Malu and Carlos Alvarez Center for Transplantation, Hepatobiliary Surgery and Innovation, The University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, San Antonio, TX, 78229, USA
- Department of Neurology, University of Texas, San Antonio, TX, 78229, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, 02139, USA
| | - Aly Karsan
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada
| | - Habil Zare
- Department of Cell Systems & Anatomy, The University of Texas Health Science Center, San Antonio, TX, 78229, USA.
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, San Antonio, TX, 78229, USA.
- Department of Cell Systems & Anatomy, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| |
Collapse
|
28
|
Chopra H, Annu, Shin DK, Munjal K, Priyanka, Dhama K, Emran TB. Revolutionizing clinical trials: the role of AI in accelerating medical breakthroughs. Int J Surg 2023; 109:4211-4220. [PMID: 38259001 PMCID: PMC10720846 DOI: 10.1097/js9.0000000000000705] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/13/2023] [Indexed: 01/24/2024]
Abstract
Clinical trials are the essential assessment for safe, reliable, and effective drug development. Data-related limitations, extensive manual efforts, remote patient monitoring, and the complexity of traditional clinical trials on patients drive the application of Artificial Intelligence (AI) in medical and healthcare organisations. For expeditious and streamlined clinical trials, a personalised AI solution is the best utilisation. AI provides broad utility options through structured, standardised, and digitally driven elements in medical research. The clinical trials are a time-consuming process with patient recruitment, enrolment, frequent monitoring, and medical adherence and retention. With an AI-powered tool, the automated data can be generated and managed for the trial lifecycle with all the records of the medical history of the patient as patient-centric AI. AI can intelligently interpret the data, feed downstream systems, and automatically fill out the required analysis report. This article explains how AI has revolutionised innovative ways of collecting data, biosimulation, and early disease diagnosis for clinical trials and overcomes the challenges more precisely through cost and time reduction, improved efficiency, and improved drug development research with less need for rework. The future implications of AI to accelerate clinical trials are important in medical research because of its fast output and overall utility.
Collapse
Affiliation(s)
- Hitesh Chopra
- Department of Biosciences, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai - 602105, Tamil Nadu, India
| | - Annu
- Thin Film and Materials Laboratory, School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dong K. Shin
- Thin Film and Materials Laboratory, School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Kavita Munjal
- Department of Pharmacy, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh 201303, India
| | - Priyanka
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University (GADVASU), Rampura Phul, Bathinda, Punjab
| | - Kuldeep Dhama
- Indian Veterinary Research Institute (IVRI), Izatnagar, Bareilly, Uttar Pradesh
| | - Talha B. Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International niversity, Dhaka, Bangladesh
| |
Collapse
|
29
|
Li X, Chen H. Characteristics of glucolipid metabolism and complications in novel cluster-based diabetes subgroups: a retrospective study. Lipids Health Dis 2023; 22:200. [PMID: 37990237 PMCID: PMC10662503 DOI: 10.1186/s12944-023-01953-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/19/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Glucolipid metabolism plays an important role in the occurrence and development of diabetes mellitus. However, there is limited research on the characteristics of glucolipid metabolism and complications in different subgroups of newly diagnosed diabetes. This study aimed to investigate the characteristics of glucolipid metabolism and complications in novel cluster-based diabetes subgroups and explore the contributions of different glucolipid metabolism indicators to the occurrence of complications and pancreatic function. METHODS This retrospective study included 547 newly diagnosed type 2 diabetes patients. Age, body mass index (BMI), glycated hemoglobin (HbA1C), homeostasis model assessment-2 beta-cell function (HOMA2-β), and homeostasis model assessment-2 insulin resistance (HOMA2-IR) were used as clustering variables. The participants were divided into 4 groups by k-means cluster analysis. The characteristics of glucolipid indicators and complications in each subgroup were analyzed. Regression analyses were used to evaluate the impact of glucolipid metabolism indicators on complications and pancreatic function. RESULTS Total cholesterol (TC), triglycerides (TG), triglyceride glucose index (TyG), HbA1C, fasting plasma glucose (FPG), and 2-h postprandial plasma glucose (2hPG) were higher in the severe insulin-resistant diabetes (SIRD) and severe insulin-deficient diabetes (SIDD) groups. Fasting insulin (FINS), fasting C-peptide (FCP), 2-h postprandial insulin (2hINS), 2-h postprandial C-peptide (2hCP), and the monocyte-to-high-density lipoprotein cholesterol ratio (MHR) were higher in mild obesity-related diabetes (MOD) and SIRD. 2hCP, FCP, and FINS were positively correlated with HOMA2-β, while FPG, TyG, HbA1C, and TG were negatively correlated with HOMA2-β. FINS, FPG, FCP, and HbA1C were positively correlated with HOMA2-IR, while high-density lipoprotein (HDL) was negatively correlated with HOMA2-IR. FINS (odds ratio (OR),1.043;95% confidence interval (CI) 1.006 ~ 1.081), FCP (OR,2.881;95%CI 2.041 ~ 4.066), and TyG (OR,1.649;95%CI 1.292 ~ 2.104) contributed to increase the risk of nonalcoholic fatty liver disease (NAFLD); 2hINS (OR,1.015;95%CI 1.008 ~ 1.022) contributed to increase the risk of atherosclerotic cardiovascular disease (ASCVD); FCP (OR,1.297;95%CI 1.027 ~ 1.637) significantly increased the risk of chronic kidney disease (CKD). CONCLUSIONS There were differences in the characteristics of glucolipid metabolism as well as complications among different subgroups of newly diagnosed type 2 diabetes. 2hCP, FCP, FINS, FPG, TyG, HbA1C, HDL and TG influenced the function of insulin. FINS, TyG, 2hINS, and FCP were associated with ASCVD, NAFLD, and CKD in newly diagnosed T2DM patients.
Collapse
Affiliation(s)
- Xinrong Li
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu Province, China
| | - Hui Chen
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
30
|
Бондарь ИА, Шабельникова ОЮ. [Clinical features and complication rates in type 2 diabetes mellitus clusters on five variables: glycated hemoglobin, age at diagnosis, body mass index, HOMA-IR, HOMA-B]. PROBLEMY ENDOKRINOLOGII 2023; 69:84-92. [PMID: 37968955 PMCID: PMC10680503 DOI: 10.14341/probl13259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 11/17/2023]
Abstract
BACKGROUND Diabetes mellitus (T2DM) is a serious medical and social problem. Now they are studying the possibility of a new stratification of diabetes. The possibility of cluster analysis for different durations of diabetes, in different cohorts to identify phenotypic clusters of T2DM and validation by reproducing clusters is relevant. AIM Identify clusters of type 2 diabetes mellitus in patients with different disease duration based on five variables: HbA1c, age at diagnosis, BMI, HOMA-IR, HOMA-B and study the clinical features and complication rates in each cluster in the Novosibirsk region. MATERIALS AND METHODS Cluster analysis of K-means was performed in 2131 patients with T2DM, aged 44 to 70 years, with a duration of diabetes of 6.42±5.66 years, living in the Novosibirsk region based on 5 variables - HbA1c, age at -diagnosis, BMI, HOMA-IR, HOMA-B. All patients a complete clinical and laboratory examination. The insulin resistance index in the HOMA (HOMA-IR, u) and the β-cell function assessment index (HOMA-B) were calculated using the calculator -version 2.2.3 at www.dtu.ox.ac.uk. RESULTS Cluster analysis revealed: Cluster 1 included 455 patients with preserved β-cell function (HOMA-B 82.97±23.28%), moderate insulin resistance (HOMA-IR 5.57±4.72) and higher diastolic BP; Cluster 2 in 1658 patients with reduced function of β-cells (HOMA-B 21.71±12.51%), the lowest indices of insulin resistance (HOMA-IR 3.50±2.48) and was characterized by a longer duration of diabetes, high fasting glycemia , HbA1c, higher eGFR and MAU, men compared with women had a 31% higher risk of developing diabetic neuropathy and 28% more diabetic nephropathy; Cluster 3 in 18 patients with high function of β-cells (HOMA-B 228.53±63.32%), severe insulin resistance (HOMA-IR 6.92±4.77), features were high incidence of men, shorter duration of diabetes, lower fasting glycemia and HbA1c, lower diastolic BP and eGFR, high incidence of early development of diabetic retinopathy after 4.00±3.6 years. CONCLUSION Cluster analysis in patients with different durations of diabetes mellitus confirmed the possibility of using cluster analysis to identify T2DM phenotypes in the Russian population. The clusters differed in the clinical characteristics of patients, the frequency and risk of diabetic complications. These results have potential value for early stratification of therapy.
Collapse
Affiliation(s)
- И. А. Бондарь
- Новосибирский государственный медицинский университет
| | | |
Collapse
|
31
|
Lu Y, Wang W, Liu J, Xie M, Liu Q, Li S. Vascular complications of diabetes: A narrative review. Medicine (Baltimore) 2023; 102:e35285. [PMID: 37800828 PMCID: PMC10553000 DOI: 10.1097/md.0000000000035285] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/28/2023] [Indexed: 10/07/2023] Open
Abstract
Diabetes mellitus is a complex chronic metabolic disease characterized by hyperglycemia and various complications. According to the different pathophysiological mechanisms, these complications can be classified as microvascular or macrovascular complications, which have long-term negative effects on vital organs such as the eyes, kidneys, heart, and brain, and lead to increased patient mortality. Diabetes mellitus is a major global health issue, and its incidence and prevalence have increased significantly in recent years. Moreover, the incidence is expected to continue to rise as more people adopt a Western lifestyle and diet. Thus, it is essential to understand the epidemiology, pathogenesis, risk factors, and treatment of vascular complications to aid patients in managing the disease effectively. This paper provides a comprehensive review of the literature to clarify the above content. Furthermore, this paper also delves into the correlation between novel risk factors, such as long noncoding RNAs, gut microbiota, and nonalcoholic fatty liver disease, with diabetic vascular complications.
Collapse
Affiliation(s)
- Yongxia Lu
- Department of Endocrinology and Metabolism, Chengdu Seventh People’s Hospital, Chengdu, China
| | - Wei Wang
- Department of Endocrinology and Metabolism, Chengdu Seventh People’s Hospital, Chengdu, China
| | - Jingyu Liu
- Department of Endocrinology and Metabolism, Chengdu Seventh People’s Hospital, Chengdu, China
| | - Min Xie
- Department of Cardiovascular Medicine, Chengdu Seventh People’s Hospital, Chengdu, China
| | - Qiang Liu
- Department of Endocrinology and Metabolism, Chengdu Seventh People’s Hospital, Chengdu, China
| | - Sufang Li
- Department of Endocrinology and Metabolism, Chengdu Seventh People’s Hospital, Chengdu, China
| |
Collapse
|
32
|
Misra S, Wagner R, Ozkan B, Schön M, Sevilla-Gonzalez M, Prystupa K, Wang CC, Kreienkamp RJ, Cromer SJ, Rooney MR, Duan D, Thuesen ACB, Wallace AS, Leong A, Deutsch AJ, Andersen MK, Billings LK, Eckel RH, Sheu WHH, Hansen T, Stefan N, Goodarzi MO, Ray D, Selvin E, Florez JC, Meigs JB, Udler MS. Precision subclassification of type 2 diabetes: a systematic review. COMMUNICATIONS MEDICINE 2023; 3:138. [PMID: 37798471 PMCID: PMC10556101 DOI: 10.1038/s43856-023-00360-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Heterogeneity in type 2 diabetes presentation and progression suggests that precision medicine interventions could improve clinical outcomes. We undertook a systematic review to determine whether strategies to subclassify type 2 diabetes were associated with high quality evidence, reproducible results and improved outcomes for patients. METHODS We searched PubMed and Embase for publications that used 'simple subclassification' approaches using simple categorisation of clinical characteristics, or 'complex subclassification' approaches which used machine learning or 'omics approaches in people with established type 2 diabetes. We excluded other diabetes subtypes and those predicting incident type 2 diabetes. We assessed quality, reproducibility and clinical relevance of extracted full-text articles and qualitatively synthesised a summary of subclassification approaches. RESULTS Here we show data from 51 studies that demonstrate many simple stratification approaches, but none have been replicated and many are not associated with meaningful clinical outcomes. Complex stratification was reviewed in 62 studies and produced reproducible subtypes of type 2 diabetes that are associated with outcomes. Both approaches require a higher grade of evidence but support the premise that type 2 diabetes can be subclassified into clinically meaningful subtypes. CONCLUSION Critical next steps toward clinical implementation are to test whether subtypes exist in more diverse ancestries and whether tailoring interventions to subtypes will improve outcomes.
Collapse
Affiliation(s)
- Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK.
| | - Robert Wagner
- Department of Endocrinology and Diabetology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Bige Ozkan
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Martin Schön
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Magdalena Sevilla-Gonzalez
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Katsiaryna Prystupa
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Caroline C Wang
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Raymond J Kreienkamp
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
| | - Sara J Cromer
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mary R Rooney
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Daisy Duan
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anne Cathrine Baun Thuesen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amelia S Wallace
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Aaron Leong
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, 100 Cambridge St 16th Floor, Boston, MA, USA
| | - Aaron J Deutsch
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mette K Andersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Liana K Billings
- Division of Endocrinology, Diabetes and Metabolism, NorthShore University Health System, Skokie, IL, USA
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Robert H Eckel
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wayne Huey-Herng Sheu
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Miaoli County, Taiwan, ROC
- Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- Division of Endocrinology and Metabolism, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Norbert Stefan
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- University Hospital of Tübingen, Tübingen, Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM), Helmholtz Center Munich, Neuherberg, Germany
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Debashree Ray
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elizabeth Selvin
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jose C Florez
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - James B Meigs
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, 100 Cambridge St 16th Floor, Boston, MA, USA
| | - Miriam S Udler
- Programs in Metabolism and Medical & Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Division of Endocrinology, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
33
|
Srivastava S, Rajan V. ExpertNet: A Deep Learning Approach to Combined Risk Modeling and Subtyping in Intensive Care Units. IEEE J Biomed Health Inform 2023; 27:5076-5086. [PMID: 37819834 DOI: 10.1109/jbhi.2023.3295751] [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: 10/13/2023]
Abstract
Risk models play a crucial role in disease prevention, particularly in intensive care units (ICUs). Diseases often have complex manifestations with heterogeneous subpopulations, or subtypes, that exhibit distinct clinical characteristics. Risk models that explicitly model subtypes have high predictive accuracy and facilitate subtype-specific personalization. Such models combine clustering and classification methods but do not effectively utilize the inferred subtypes in risk modeling. Their limitations include tendency to obtain degenerate clusters and cluster-specific data scarcity leading to insufficient training data for the corresponding classifier. In this article, we develop a new deep learning model for simultaneous clustering and classification, ExpertNet, with novel loss terms and network training strategies that address these limitations. The performance of ExpertNet is evaluated on the tasks of predicting risk of (i) sepsis and (ii) acute respiratory distress syndrome (ARDS), using two large electronic medical records datasets from ICUs. Our extensive experiments show that, in comparison to state-of-the-art baselines for combined clustering and classification, ExpertNet achieves superior accuracy in risk prediction for both ARDS and sepsis; and comparable clustering performance. Visual analysis of the clusters further demonstrates that the clusters obtained are clinically meaningful and a knowledge-distilled model shows significant differences in risk factors across the subtypes. By addressing technical challenges in training neural networks for simultaneous clustering and classification, ExpertNet lays the algorithmic foundation for the future development of subtype-aware risk models.
Collapse
|
34
|
Shen Y, Gao Y, Shi J, Huang Z, Dai R, Fu Y, Zhou Y, Kong W, Cui Q. MicroRNA-disease Network Analysis Repurposes Methotrexate for the Treatment of Abdominal Aortic Aneurysm in Mice. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:1030-1042. [PMID: 36030000 PMCID: PMC10928436 DOI: 10.1016/j.gpb.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 07/15/2022] [Accepted: 08/19/2022] [Indexed: 06/15/2023]
Abstract
Abdominal aortic aneurysm (AAA) is a permanent dilatation of the abdominal aorta and is highly lethal. The main purpose of the current study is to search for noninvasive medical therapies for AAA, for which there is currently no effective drug therapy. Network medicine represents a cutting-edge technology, as analysis and modeling of disease networks can provide critical clues regarding the etiology of specific diseases and therapeutics that may be effective. Here, we proposed a novel algorithm to quantify disease relations based on a large accumulated microRNA-disease association dataset and then built a disease network covering 15 disease classes and 304 diseases. Analysis revealed some patterns for these diseases. For instance, diseases tended to be clustered and coherent in the network. Surprisingly, we found that AAA showed the strongest similarity with rheumatoid arthritis and systemic lupus erythematosus, both of which are autoimmune diseases, suggesting that AAA could be one type of autoimmune diseases in etiology. Based on this observation, we further hypothesized that drugs for autoimmune diseases could be repurposed for the prevention and therapy of AAA. Finally, animal experiments confirmed that methotrexate, a drug for autoimmune diseases, was able to alleviate the formation and development of AAA.
Collapse
Affiliation(s)
- Yicong Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yuanxu Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; State Key Laboratory of Lunar and Planetary Sciences, Macau University of Science and Technology, Macao Special Administrative Region 999078, China; Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Jiangcheng Shi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Zhou Huang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Rongbo Dai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yuan Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China.
| | - Qinghua Cui
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Biomedical Informatics, Center for Noncoding RNA Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
35
|
Paranjpe I, Wang X, Anandakrishnan N, Haydak JC, Van Vleck T, DeFronzo S, Li Z, Mendoza A, Liu R, Fu J, Forrest I, Zhou W, Lee K, O'Hagan R, Dellepiane S, Menon KM, Gulamali F, Kamat S, Gusella GL, Charney AW, Hofer I, Cho JH, Do R, Glicksberg BS, He JC, Nadkarni GN, Azeloglu EU. Deep learning on electronic medical records identifies distinct subphenotypes of diabetic kidney disease driven by genetic variations in the Rho pathway. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.06.23295120. [PMID: 37732187 PMCID: PMC10508814 DOI: 10.1101/2023.09.06.23295120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Kidney disease affects 50% of all diabetic patients; however, prediction of disease progression has been challenging due to inherent disease heterogeneity. We use deep learning to identify novel genetic signatures prognostically associated with outcomes. Using autoencoders and unsupervised clustering of electronic health record data on 1,372 diabetic kidney disease patients, we establish two clusters with differential prevalence of end-stage kidney disease. Exome-wide associations identify a novel variant in ARHGEF18, a Rho guanine exchange factor specifically expressed in glomeruli. Overexpression of ARHGEF18 in human podocytes leads to impairments in focal adhesion architecture, cytoskeletal dynamics, cellular motility, and RhoA/Rac1 activation. Mutant GEF18 is resistant to ubiquitin mediated degradation leading to pathologically increased protein levels. Our findings uncover the first known disease-causing genetic variant that affects protein stability of a cytoskeletal regulator through impaired degradation, a potentially novel class of expression quantitative trait loci that can be therapeutically targeted.
Collapse
|
36
|
Yu A, Zhong Y, Feng X, Wei Y. Quantile regression for nonignorable missing data with its application of analyzing electronic medical records. Biometrics 2023; 79:2036-2049. [PMID: 35861675 DOI: 10.1111/biom.13723] [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: 11/18/2021] [Accepted: 07/15/2022] [Indexed: 11/27/2022]
Abstract
Over the past decade, there has been growing enthusiasm for using electronic medical records (EMRs) for biomedical research. Quantile regression estimates distributional associations, providing unique insights into the intricacies and heterogeneity of the EMR data. However, the widespread nonignorable missing observations in EMR often obscure the true associations and challenge its potential for robust biomedical discoveries. We propose a novel method to estimate the covariate effects in the presence of nonignorable missing responses under quantile regression. This method imposes no parametric specifications on response distributions, which subtly uses implicit distributions induced by the corresponding quantile regression models. We show that the proposed estimator is consistent and asymptotically normal. We also provide an efficient algorithm to obtain the proposed estimate and a randomly weighted bootstrap approach for statistical inferences. Numerical studies, including an empirical analysis of real-world EMR data, are used to assess the proposed method's finite-sample performance compared to existing literature.
Collapse
Affiliation(s)
- Aiai Yu
- School of Statistics and Management, Shanghai University of Finance and Economics, Shanghai, China
| | - Yujie Zhong
- School of Statistics and Management, Shanghai University of Finance and Economics, Shanghai, China
| | - Xingdong Feng
- School of Statistics and Management, Shanghai University of Finance and Economics, Shanghai, China
| | - Ying Wei
- Department of Biostatistics, Columbia University, New York, New York, USA
| |
Collapse
|
37
|
Sajedi S, Ebrahimi G, Roudi R, Mehta I, Samimi H, Kazempour S, Zainulabadeen A, Docking TR, Arora SP, Cigarroa F, Seshadri S, Karsan A, Zare H. "iNETgrate": integrating DNA methylation and gene expression data in a single gene network. RESEARCH SQUARE 2023:rs.3.rs-3246325. [PMID: 37645739 PMCID: PMC10462231 DOI: 10.21203/rs.3.rs-3246325/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Integrating multi-omics data in one model can increase statistical power. However, designing such a model is challenging because different omics are measured at different levels. We developed the iNETgrate package (https://bioconductor.org/packages/iNETgrate/) that efficiently integrates transcriptome and DNA methylation data in a single gene network. Applying iNETgrate on five independent datasets improved prognostication compared to common clinical gold standards and a patient similarity network approach.
Collapse
Affiliation(s)
- Sogand Sajedi
- Department of Cell Systems & Anatomy, The University of Texas Health Science Center, San Antonio, Texas 78229, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, San Antonio, Texas 78229, USA
| | - Ghazal Ebrahimi
- Bioinformatics Program, the University of British Columbia, Vancouver, BC, Canada
| | - Raheleh Roudi
- Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Isha Mehta
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Hanie Samimi
- School of Architecture, University of Utah, Salt Lake City, Utah 84112, USA
| | - Shiva Kazempour
- Department of Cell Systems & Anatomy, The University of Texas Health Science Center, San Antonio, Texas 78229, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, San Antonio, Texas 78229, USA
| | - Aamir Zainulabadeen
- Department of Computer Science, Princeton University, Princeton, New Jersey 08540, USA
| | - Thomas Roderick Docking
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Research Centre, Vancouver, British Columbia, V5Z 1L3, Canada
| | - Sukeshi Patel Arora
- Mays Cancer Center, The University of Texas Health Science Center, San Antonio, Texas 78229, USA
| | - Francisco Cigarroa
- Malu and Carlos Alvarez Center for Transplantation, Hepatobiliary Surgery and Innovation, The University of Texas Health Science Center, San Antonio, Texas 78229, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, San Antonio, Texas 78229, USA
- Department of Neurology, University of Texas, San Antonio, Texas 78229, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02139,USA
| | - Aly Karsan
- Canada’s Michael Smith Genome Sciences Centre, British Columbia Cancer Research Centre, Vancouver, British Columbia, V5Z 1L3, Canada
| | - Habil Zare
- Department of Cell Systems & Anatomy, The University of Texas Health Science Center, San Antonio, Texas 78229, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, San Antonio, Texas 78229, USA
| |
Collapse
|
38
|
Komuro J, Kusumoto D, Hashimoto H, Yuasa S. Machine learning in cardiology: Clinical application and basic research. J Cardiol 2023; 82:128-133. [PMID: 37141938 DOI: 10.1016/j.jjcc.2023.04.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/06/2023]
Abstract
Machine learning is a subfield of artificial intelligence. The quality and versatility of machine learning have been rapidly improving and playing a critical role in many aspects of social life. This trend is also observed in the medical field. Generally, there are three main types of machine learning: supervised, unsupervised, and reinforcement learning. Each type of learning is adequately selected for the purpose and type of data. In the field of medicine, various types of information are collected and used, and research using machine learning is becoming increasingly relevant. Many clinical studies are conducted using electronic health and medical records, including in the cardiovascular area. Machine learning has also been applied in basic research. Machine learning has been widely used for several types of data analysis, such as clustering of microarray analysis and RNA sequence analysis. Machine learning is essential for genome and multi-omics analyses. This review summarizes the recent advancements in the use of machine learning in clinical applications and basic cardiovascular research.
Collapse
Affiliation(s)
- Jin Komuro
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Dai Kusumoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Hisayuki Hashimoto
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Shinsuke Yuasa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
39
|
Duman AN, Tatar AE. Topological data analysis for revealing dynamic brain reconfiguration in MEG data. PeerJ 2023; 11:e15721. [PMID: 37489123 PMCID: PMC10363343 DOI: 10.7717/peerj.15721] [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: 01/13/2023] [Accepted: 06/16/2023] [Indexed: 07/26/2023] Open
Abstract
In recent years, the focus of the functional connectivity community has shifted from stationary approaches to the ones that include temporal dynamics. Especially, non-invasive electrophysiological data (magnetoencephalography/electroencephalography (MEG/EEG)) with high temporal resolution and good spatial coverage have made it possible to measure the fast alterations in the neural activity in the brain during ongoing cognition. In this article, we analyze dynamic brain reconfiguration using MEG images collected from subjects during the rest and the cognitive tasks. Our proposed topological data analysis method, called Mapper, produces biomarkers that differentiate cognitive tasks without prior spatial and temporal collapse of the data. The suggested method provides an interactive visualization of the rapid fluctuations in electrophysiological data during motor and cognitive tasks; hence, it has the potential to extract clinically relevant information at an individual level without temporal and spatial collapse.
Collapse
Affiliation(s)
- Ali Nabi Duman
- Department of Mathematics, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Ahmet E. Tatar
- Center for Information Technology, University of Groningen, Groningen, Netherlands
| |
Collapse
|
40
|
Wamil M, Hassaine A, Rao S, Li Y, Mamouei M, Canoy D, Nazarzadeh M, Bidel Z, Copland E, Rahimi K, Salimi-Khorshidi G. Stratification of diabetes in the context of comorbidities, using representation learning and topological data analysis. Sci Rep 2023; 13:11478. [PMID: 37455284 PMCID: PMC10350454 DOI: 10.1038/s41598-023-38251-1] [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: 12/15/2022] [Accepted: 07/05/2023] [Indexed: 07/18/2023] Open
Abstract
Diabetes is a heterogenous, multimorbid disorder with a large variation in manifestations, trajectories, and outcomes. The aim of this study is to validate a novel machine learning method for the phenotyping of diabetes in the context of comorbidities. Data from 9967 multimorbid patients with a new diagnosis of diabetes were extracted from Clinical Practice Research Datalink. First, using BEHRT (a transformer-based deep learning architecture), the embeddings corresponding to diabetes were learned. Next, topological data analysis (TDA) was carried out to test how different areas in high-dimensional manifold correspond to different risk profiles. The following endpoints were considered when profiling risk trajectories: major adverse cardiovascular events (MACE), coronary artery disease (CAD), stroke (CVA), heart failure (HF), renal failure (RF), diabetic neuropathy, peripheral arterial disease, reduced visual acuity and all-cause mortality. Kaplan Meier curves were plotted for each derived phenotype. Finally, we tested the performance of an established risk prediction model (QRISK) by adding TDA-derived features. We identified four subgroups of patients with diabetes and divergent comorbidity patterns differing in their risk of future cardiovascular, renal, and other microvascular outcomes. Phenotype 1 (young with chronic inflammatory conditions) and phenotype 2 (young with CAD) included relatively younger patients with diabetes compared to phenotypes 3 (older with hypertension and renal disease) and 4 (older with previous CVA), and those subgroups had a higher frequency of pre-existing cardio-renal diseases. Within ten years of follow-up, 2592 patients (26%) experienced MACE, 2515 patients (25%) died, and 2020 patients (20%) suffered RF. QRISK3 model's AUC was augmented from 67.26% (CI 67.25-67.28%) to 67.67% (CI 67.66-67.69%) by adding specific TDA-derived phenotype and the distances to both extremities of the TDA graph improving its performance in the prediction of CV outcomes. We confirmed the importance of accounting for multimorbidity when risk stratifying heterogenous cohort of patients with new diagnosis of diabetes. Our unsupervised machine learning method improved the prediction of clinical outcomes.
Collapse
Affiliation(s)
- Malgorzata Wamil
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK.
- Mayo Clinic Healthcare, 15 Portland Place, London, UK.
| | - Abdelaali Hassaine
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Science Division, University of Oxford, Oxford, UK
| | - Shishir Rao
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Science Division, University of Oxford, Oxford, UK
| | - Yikuan Li
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Science Division, University of Oxford, Oxford, UK
| | - Mohammad Mamouei
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Science Division, University of Oxford, Oxford, UK
| | - Dexter Canoy
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Science Division, University of Oxford, Oxford, UK
| | - Milad Nazarzadeh
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Science Division, University of Oxford, Oxford, UK
| | - Zeinab Bidel
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Science Division, University of Oxford, Oxford, UK
| | - Emma Copland
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Science Division, University of Oxford, Oxford, UK
| | - Kazem Rahimi
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Science Division, University of Oxford, Oxford, UK
| | - Gholamreza Salimi-Khorshidi
- Deep Medicine, Oxford Martin School, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Science Division, University of Oxford, Oxford, UK
| |
Collapse
|
41
|
Zhao S, Li H, Jing X, Zhang X, Li R, Li Y, Liu C, Chen J, Li G, Zheng W, Li Q, Wang X, Wang L, Sun Y, Xu Y, Wang S. Identifying subgroups of patients with type 2 diabetes based on real-world traditional chinese medicine electronic medical records. Front Pharmacol 2023; 14:1210667. [PMID: 37456755 PMCID: PMC10339739 DOI: 10.3389/fphar.2023.1210667] [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: 04/23/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction: Type 2 diabetes (T2D) is a multifactorial complex chronic disease with a high prevalence worldwide, and Type 2 diabetes patients with different comorbidities often present multiple phenotypes in the clinic. Thus, there is a pressing need to improve understanding of the complexity of the clinical Type 2 diabetes population to help identify more accurate disease subtypes for personalized treatment. Methods: Here, utilizing the traditional Chinese medicine (TCM) clinical electronic medical records (EMRs) of 2137 Type 2 diabetes inpatients, we followed a heterogeneous medical record network (HEMnet) framework to construct heterogeneous medical record networks by integrating the clinical features from the electronic medical records, molecular interaction networks and domain knowledge. Results: Of the 2137 Type 2 diabetes patients, 1347 were male (63.03%), and 790 were female (36.97%). Using the HEMnet method, we obtained eight non-overlapping patient subgroups. For example, in H3, Poria, Astragali Radix, Glycyrrhizae Radix et Rhizoma, Cinnamomi Ramulus, and Liriopes Radix were identified as significant botanical drugs. Cardiovascular diseases (CVDs) were found to be significant comorbidities. Furthermore, enrichment analysis showed that there were six overlapping pathways and eight overlapping Gene Ontology terms among the herbs, comorbidities, and Type 2 diabetes in H3. Discussion: Our results demonstrate that identification of the Type 2 diabetes subgroup based on the HEMnet method can provide important guidance for the clinical use of herbal prescriptions and that this method can be used for other complex diseases.
Collapse
Affiliation(s)
- Shuai Zhao
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hengfei Li
- Department of Infectious Diseases, Hubei Provincial Hospital of Traditional Chinese Medicine (Affiliated Hospital of Hubei University of Chinese Medicine, Hubei Province Academy of Traditional Chinese Medicine), Wuhan, China
| | - Xuan Jing
- Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, China
| | - Xuebin Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ronghua Li
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yinghao Li
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chenguang Liu
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Chen
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoxia Li
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfei Zheng
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Li
- Department of Nursing, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xue Wang
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Letian Wang
- Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuanyuan Sun
- Department of Obstetrics and Gynecology, Weifang Fangzi District People’s Hospital, Weifang, China
| | - Yunsheng Xu
- Department of Endocrinology, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shihua Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Khan SR, Rost H, Cox B, Razani B, Alexeeff S, Wheeler MB, Gunderson EP. Heterogeneity in Early Postpartum Metabolic Profiles Among Women with GDM Who Progressed to Type 2 Diabetes During 10-Year Follow-Up: The SWIFT Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.13.23291346. [PMID: 37398098 PMCID: PMC10312884 DOI: 10.1101/2023.06.13.23291346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
GDM is a strong risk factor for progression to T2D after pregnancy. Although both GDM and T2D exhibit heterogeneity, the link between the distinct heterogeneity of GDM and incident T2D has not been established. Herein, we evaluate early postpartum profiles of women with recent GDM who later developed incident T2D using a soft clustering method, followed by the integration of both clinical phenotypic variables and metabolomics to characterize these heterogeneous clusters/groups clinically and their molecular mechanisms. We identified three clusters based on two indices of glucose homeostasis at 6-9 weeks postpartum - HOMA-IR and HOMA-B among women who developed incident T2D during the 12-year follow-up. The clusters were classified as follows: pancreatic beta-cell dysfunction group (cluster-1), insulin resistant group (cluster-3), and a combination of both phenomena (cluster-2) comprising the majority of T2D. We also identified postnatal blood test parameters to distinguish the three clusters for clinical testing. Moreover, we compared these three clusters in their metabolomics profiles at the early stage of the disease to identify the mechanistic insights. A significantly higher concentration of a metabolite at the early stage of a T2D cluster than other clusters indicates its essentiality for the particular disease character. As such, the early-stage characters of T2D cluster-1 pathology include a higher concentration of sphingolipids, acyl-alkyl phosphatidylcholines, lysophosphatidylcholines, and glycine, indicating their essentiality for pancreatic beta-cell function. In contrast, the early-stage characteristics of T2D cluster-3 pathology include a higher concentration of diacyl phosphatidylcholines, acyl-carnitines, isoleucine, and glutamate, indicating their essentiality for insulin actions. Notably, all these biomolecules are found in the T2D cluster-2 with mediocre concentrations, indicating a true nature of a mixed group. In conclusion, we have deconstructed incident T2D heterogeneity and identified three clusters with their clinical testing procedures and molecular mechanisms. This information will aid in adopting proper interventions using a precision medicine approach.
Collapse
Affiliation(s)
- Saifur R Khan
- Department of Cardiology, University of Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, PA, USA
- Departments of Physiology and Medicine, University of Toronto, Ontario, Canada
| | - Hannes Rost
- Donnelly Centre, University of Toronto, Ontario, Canada
| | - Brian Cox
- Department of Obstetrics and Gynaecology, University of Toronto, Ontario, Canada
| | - Babak Razani
- Department of Cardiology, University of Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, PA, USA
| | - Stacey Alexeeff
- Kaiser Permanente Northern California, Division of Research, Oakland, CA
| | - Michael B Wheeler
- Departments of Physiology and Medicine, University of Toronto, Ontario, Canada
| | - Erica P Gunderson
- Kaiser Permanente Northern California, Division of Research, Oakland, CA
- Kaiser Permanente Bernard J. Tyson School of Medicine, Department of Health Systems Science, Pasadena, CA
| |
Collapse
|
43
|
Ratter-Rieck JM, Roden M, Herder C. Diabetes and climate change: current evidence and implications for people with diabetes, clinicians and policy stakeholders. Diabetologia 2023; 66:1003-1015. [PMID: 36964771 PMCID: PMC10039694 DOI: 10.1007/s00125-023-05901-y] [Citation(s) in RCA: 16] [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: 01/19/2023] [Accepted: 02/15/2023] [Indexed: 03/26/2023]
Abstract
Climate change will be a major challenge for the world's health systems in the coming decades. Elevated temperatures and increasing frequencies of heat waves, wildfires, heavy precipitation and other weather extremes can affect health in many ways, especially if chronic diseases are already present. Impaired responses to heat stress, including compromised vasodilation and sweating, diabetes-related comorbidities, insulin resistance and chronic low-grade inflammation make people with diabetes particularly vulnerable to environmental risk factors, such as extreme weather events and air pollution. Additionally, multiple pathogens show an increased rate of transmission under conditions of climate change and people with diabetes have an altered immune system, which increases the risk for a worse course of infectious diseases. In this review, we summarise recent studies on the impact of climate-change-associated risk for people with diabetes and discuss which individuals may be specifically prone to these risk conditions due to their clinical features. Knowledge of such high-risk groups will help to develop and implement tailored prevention and management strategies to mitigate the detrimental effect of climate change on the health of people with diabetes.
Collapse
Affiliation(s)
- Jacqueline M Ratter-Rieck
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany.
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
44
|
Ledziński Ł, Grześk G. Artificial Intelligence Technologies in Cardiology. J Cardiovasc Dev Dis 2023; 10:jcdd10050202. [PMID: 37233169 DOI: 10.3390/jcdd10050202] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
As the world produces exabytes of data, there is a growing need to find new methods that are more suitable for dealing with complex datasets. Artificial intelligence (AI) has significant potential to impact the healthcare industry, which is already on the road to change with the digital transformation of vast quantities of information. The implementation of AI has already achieved success in the domains of molecular chemistry and drug discoveries. The reduction in costs and in the time needed for experiments to predict the pharmacological activities of new molecules is a milestone in science. These successful applications of AI algorithms provide hope for a revolution in healthcare systems. A significant part of artificial intelligence is machine learning (ML), of which there are three main types-supervised learning, unsupervised learning, and reinforcement learning. In this review, the full scope of the AI workflow is presented, with explanations of the most-often-used ML algorithms and descriptions of performance metrics for both regression and classification. A brief introduction to explainable artificial intelligence (XAI) is provided, with examples of technologies that have developed for XAI. We review important AI implementations in cardiology for supervised, unsupervised, and reinforcement learning and natural language processing, emphasizing the used algorithm. Finally, we discuss the need to establish legal, ethical, and methodical requirements for the deployment of AI models in medicine.
Collapse
Affiliation(s)
- Łukasz Ledziński
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Ujejskiego 75, 85-168 Bydgoszcz, Poland
| | - Grzegorz Grześk
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Ujejskiego 75, 85-168 Bydgoszcz, Poland
| |
Collapse
|
45
|
Jia G, Li Y, Zhong X, Wang K, Pividori M, Alomairy R, Esposito A, Ltaief H, Terao C, Akiyama M, Matsuda K, Keyes DE, Im HK, Gojobori T, Kamatani Y, Kubo M, Cox NJ, Evans J, Gao X, Rzhetsky A. The high-dimensional space of human diseases built from diagnosis records and mapped to genetic loci. NATURE COMPUTATIONAL SCIENCE 2023; 3:403-417. [PMID: 38177845 PMCID: PMC10766526 DOI: 10.1038/s43588-023-00453-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 04/13/2023] [Indexed: 01/06/2024]
Abstract
Human diseases are traditionally studied as singular, independent entities, limiting researchers' capacity to view human illnesses as dependent states in a complex, homeostatic system. Here, using time-stamped clinical records of over 151 million unique Americans, we construct a disease representation as points in a continuous, high-dimensional space, where diseases with similar etiology and manifestations lie near one another. We use the UK Biobank cohort, with half a million participants, to perform a genome-wide association study of newly defined human quantitative traits reflecting individuals' health states, corresponding to patient positions in our disease space. We discover 116 genetic associations involving 108 genetic loci and then use ten disease constellations resulting from clustering analysis of diseases in the embedding space, as well as 30 common diseases, to demonstrate that these genetic associations can be used to robustly predict various morbidities.
Collapse
Affiliation(s)
- Gengjie Jia
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Yu Li
- Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Xue Zhong
- Department of Medicine and Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, US
| | - Kanix Wang
- Department of Medicine, Institute of Genomics and Systems Biology, Committee on Genomics, Genetics, and Systems Biology, University of Chicago, Chicago, IL, US
- Department of Operations, Business Analytics, and Information Systems, University of Cincinnati, Cincinnati, OH, US
| | - Milton Pividori
- Department of Medicine, Institute of Genomics and Systems Biology, Committee on Genomics, Genetics, and Systems Biology, University of Chicago, Chicago, IL, US
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, US
| | - Rabab Alomairy
- Extreme Computing Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
- College of Computer Science and Engineering, University of Jeddah, Jeddah, Saudi Arabia
| | | | - Hatem Ltaief
- Extreme Computing Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Chikashi Terao
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Department of Applied Genetics, The School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Masato Akiyama
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Matsuda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - David E Keyes
- Extreme Computing Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Hae Kyung Im
- Department of Medicine, Institute of Genomics and Systems Biology, Committee on Genomics, Genetics, and Systems Biology, University of Chicago, Chicago, IL, US
| | - Takashi Gojobori
- Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Yoichiro Kamatani
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Nancy J Cox
- Department of Medicine and Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, US
| | - James Evans
- Department of Sociology, University of Chicago, Chicago, IL, US
| | - Xin Gao
- Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.
| | - Andrey Rzhetsky
- Department of Medicine, Institute of Genomics and Systems Biology, Committee on Genomics, Genetics, and Systems Biology, University of Chicago, Chicago, IL, US.
- Department of Human Genetics, University of Chicago, Chicago, IL, US.
| |
Collapse
|
46
|
Amézquita EJ, Nasrin F, Storey KM, Yoshizawa M. Genomics data analysis via spectral shape and topology. PLoS One 2023; 18:e0284820. [PMID: 37099525 PMCID: PMC10132553 DOI: 10.1371/journal.pone.0284820] [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: 10/29/2022] [Accepted: 04/09/2023] [Indexed: 04/27/2023] Open
Abstract
Mapper, a topological algorithm, is frequently used as an exploratory tool to build a graphical representation of data. This representation can help to gain a better understanding of the intrinsic shape of high-dimensional genomic data and to retain information that may be lost using standard dimension-reduction algorithms. We propose a novel workflow to process and analyze RNA-seq data from tumor and healthy subjects integrating Mapper, differential gene expression, and spectral shape analysis. Precisely, we show that a Gaussian mixture approximation method can be used to produce graphical structures that successfully separate tumor and healthy subjects, and produce two subgroups of tumor subjects. A further analysis using DESeq2, a popular tool for the detection of differentially expressed genes, shows that these two subgroups of tumor cells bear two distinct gene regulations, suggesting two discrete paths for forming lung cancer, which could not be highlighted by other popular clustering methods, including t-distributed stochastic neighbor embedding (t-SNE). Although Mapper shows promise in analyzing high-dimensional data, tools to statistically analyze Mapper graphical structures are limited in the existing literature. In this paper, we develop a scoring method using heat kernel signatures that provides an empirical setting for statistical inferences such as hypothesis testing, sensitivity analysis, and correlation analysis.
Collapse
Affiliation(s)
- Erik J. Amézquita
- Computational Mathematics, Science, and Engineering, Michigan State University, East Lansing, MI, United States of America
| | - Farzana Nasrin
- Department of Mathematics, University of Hawaii at Manoa, Honolulu, HI, United States of America
| | - Kathleen M. Storey
- Department of Mathematics, Lafayette College, Easton, PA, United States of America
| | - Masato Yoshizawa
- School of Life Sciences, University of Hawaii at Manoa, Honolulu, HI, United States of America
| |
Collapse
|
47
|
Forrest IS, Petrazzini BO, Duffy Á, Park JK, O'Neal AJ, Jordan DM, Rocheleau G, Nadkarni GN, Cho JH, Blazer AD, Do R. A machine learning model identifies patients in need of autoimmune disease testing using electronic health records. Nat Commun 2023; 14:2385. [PMID: 37169741 PMCID: PMC10130143 DOI: 10.1038/s41467-023-37996-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 04/05/2023] [Indexed: 05/13/2023] Open
Abstract
Systemic autoimmune rheumatic diseases (SARDs) can lead to irreversible damage if left untreated, yet these patients often endure long diagnostic journeys before being diagnosed and treated. Machine learning may help overcome the challenges of diagnosing SARDs and inform clinical decision-making. Here, we developed and tested a machine learning model to identify patients who should receive rheumatological evaluation for SARDs using longitudinal electronic health records of 161,584 individuals from two institutions. The model demonstrated high performance for predicting cases of autoantibody-tested individuals in a validation set, an external test set, and an independent cohort with a broader case definition. This approach identified more individuals for autoantibody testing compared with current clinical standards and a greater proportion of autoantibody carriers among those tested. Diagnoses of SARDs and other autoimmune conditions increased with higher model probabilities. The model detected a need for autoantibody testing and rheumatology encounters up to five years before the test date and assessment date, respectively. Altogether, these findings illustrate that the clinical manifestations of a diverse array of autoimmune conditions are detectable in electronic health records using machine learning, which may help systematize and accelerate autoimmune testing.
Collapse
Affiliation(s)
- Iain S Forrest
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The BioMe Phenomics Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ben O Petrazzini
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Áine Duffy
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joshua K Park
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anya J O'Neal
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Daniel M Jordan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ghislain Rocheleau
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The BioMe Phenomics Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Judy H Cho
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The BioMe Phenomics Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashira D Blazer
- Division of Rheumatology, Hospital for Special Surgery, New York, NY, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The BioMe Phenomics Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
48
|
Misra S, Wagner R, Ozkan B, Schön M, Sevilla-Gonzalez M, Prystupa K, Wang CC, Kreienkamp RJ, Cromer SJ, Rooney MR, Duan D, Thuesen ACB, Wallace AS, Leong A, Deutsch AJ, Andersen MK, Billings LK, Eckel RH, Sheu WHH, Hansen T, Stefan N, Goodarzi MO, Ray D, Selvin E, Florez JC, Meigs JB, Udler MS. Systematic review of precision subclassification of type 2 diabetes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.19.23288577. [PMID: 37131632 PMCID: PMC10153304 DOI: 10.1101/2023.04.19.23288577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Heterogeneity in type 2 diabetes presentation, progression and treatment has the potential for precision medicine interventions that can enhance care and outcomes for affected individuals. We undertook a systematic review to ascertain whether strategies to subclassify type 2 diabetes are associated with improved clinical outcomes, show reproducibility and have high quality evidence. We reviewed publications that deployed 'simple subclassification' using clinical features, biomarkers, imaging or other routinely available parameters or 'complex subclassification' approaches that used machine learning and/or genomic data. We found that simple stratification approaches, for example, stratification based on age, body mass index or lipid profiles, had been widely used, but no strategy had been replicated and many lacked association with meaningful outcomes. Complex stratification using clustering of simple clinical data with and without genetic data did show reproducible subtypes of diabetes that had been associated with outcomes such as cardiovascular disease and/or mortality. Both approaches require a higher grade of evidence but support the premise that type 2 diabetes can be subclassified into meaningful groups. More studies are needed to test these subclassifications in more diverse ancestries and prove that they are amenable to interventions.
Collapse
|
49
|
Hwang YC, Ahn HY, Jun JE, Jeong IK, Ahn KJ, Chung HY. Subtypes of type 2 diabetes and their association with outcomes in Korean adults - A cluster analysis of community-based prospective cohort. Metabolism 2023; 141:155514. [PMID: 36746321 DOI: 10.1016/j.metabol.2023.155514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/06/2023]
Abstract
BACKGROUND Little is known about the subtypes of type 2 diabetes (T2D) and their association with clinical outcomes in Asians. METHODS We performed data-driven cluster analysis in patients with newly diagnosed drug-naive T2D (n = 756) from the Korean Genome and Epidemiology Study. Clusters were based on five variables (age at diagnosis, BMI, HbA1c, and HOMA2 β-cell function, and insulin resistance). RESULTS We identified four clusters of patients with T2D according to k-means clustering: cluster 1 (22.4 %, severe insulin-resistant diabetes [SIRD]), cluster 2 (32.7 %, mild age-related diabetes [MARD]), cluster 3 (32.7 %, mild obesity-related diabetes [MOD]), and cluster 4 (12.3 %, severe insulin-deficient diabetes [SIDD]). During 14 years of follow-up, individuals in the SIDD cluster had the highest risk of initiation of glucose-lowering therapy compared to individuals in the other three clusters. Individuals in the MARD and SIDD clusters showed the highest risk of chronic kidney disease and cardiovascular disease, and individuals in the MOD clusters showed the lowest risk after adjusting for other risk factors (P < 0.05). CONCLUSIONS Patients with T2D can be categorized into four subgroups with different glycemic deterioration and risks of diabetes complications. Individualized management might be helpful for better clinical outcomes in Asian patients with different T2D subgroups.
Collapse
Affiliation(s)
- You-Cheol Hwang
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea.
| | - Hong-Yup Ahn
- Department of Statistics, Dongguk University, Seoul, Republic of Korea
| | - Ji Eun Jun
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - In-Kyung Jeong
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Kyu Jeung Ahn
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Ho Yeon Chung
- Division of Endocrinology and Metabolism, Department of Medicine, Kyung Hee University School of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| |
Collapse
|
50
|
Kresoja KP, Unterhuber M, Wachter R, Thiele H, Lurz P. A cardiologist's guide to machine learning in cardiovascular disease prognosis prediction. Basic Res Cardiol 2023; 118:10. [PMID: 36939941 PMCID: PMC10027799 DOI: 10.1007/s00395-023-00982-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/21/2023] [Accepted: 02/26/2023] [Indexed: 03/21/2023]
Abstract
A modern-day physician is faced with a vast abundance of clinical and scientific data, by far surpassing the capabilities of the human mind. Until the last decade, advances in data availability have not been accompanied by analytical approaches. The advent of machine learning (ML) algorithms might improve the interpretation of complex data and should help to translate the near endless amount of data into clinical decision-making. ML has become part of our everyday practice and might even further change modern-day medicine. It is important to acknowledge the role of ML in prognosis prediction of cardiovascular disease. The present review aims on preparing the modern physician and researcher for the challenges that ML might bring, explaining basic concepts but also caveats that might arise when using these methods. Further, a brief overview of current established classical and emerging concepts of ML disease prediction in the fields of omics, imaging and basic science is presented.
Collapse
Affiliation(s)
- Karl-Patrik Kresoja
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Struempellstr. 39, 04289, Leipzig, Germany
- Leipzig Heart Institute, Leipzig Heart Science at Heart Center Leipzig, Leipzig, Germany
| | - Matthias Unterhuber
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Struempellstr. 39, 04289, Leipzig, Germany
- Leipzig Heart Institute, Leipzig Heart Science at Heart Center Leipzig, Leipzig, Germany
| | - Rolf Wachter
- Department of Cardiology, University Hospital Leipzig, Leipzig, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen, Göttingen, Germany
- German Cardiovascular Research Center (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Holger Thiele
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Struempellstr. 39, 04289, Leipzig, Germany.
- Leipzig Heart Institute, Leipzig Heart Science at Heart Center Leipzig, Leipzig, Germany.
| | - Philipp Lurz
- Department of Internal Medicine/Cardiology, Heart Center Leipzig at University of Leipzig, Struempellstr. 39, 04289, Leipzig, Germany.
- Leipzig Heart Institute, Leipzig Heart Science at Heart Center Leipzig, Leipzig, Germany.
| |
Collapse
|