1
|
Chen S, Li Y, Zhang H, Li J, Yang L, Wang Q, Zhang S, Luo P, Wang H, Jiang H. Multilayered visual metabolomics analysis framework for enhanced exploration of functional components in wolfberry. Food Chem 2025; 477:143583. [PMID: 40023033 DOI: 10.1016/j.foodchem.2025.143583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
Wolfberry, regarded as a nutritious fruit, has garnered significant attention in the food industry due to potential health benefits. However, the tissue-specific distribution and dynamic accumulation patterns of nutritional metabolites such as flavonoids are still unclear. In this study, a novel spatial metabolomics framework was developed, incorporating instrumental optimization, metabolite identification, molecular network analysis, metabolic pathway mapping, and machine learning-based imaging. Using DESI-MSI, this approach enabled rapid, non-destructive, in situ analysis of wolfberry metabolites with enhanced sensitivity and spatial resolution. Detailed insights into chemical and spatial changes during ripening were obtained, with a focus on flavonoids. The visualization of the flavonoid biosynthetic pathway highlighted the impact of C-3 hydroxylation on flavonoid redistribution. Furthermore, a classification model achieved a prediction accuracy exceeding 99 %, consistent with metabolic network analyses. This framework provides a powerful tool for plant metabolomics, facilitating the exploration of functional components and metabolic pathways.
Collapse
Affiliation(s)
- Shiqi Chen
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, National Key Laboratory of Veterinary Public Health Security, Beijing 100193, China
| | - Yifan Li
- Sichuan Institute for Drug Control (Sichuan Testing Center of Medical Devices), NMAP Key Laboratory of Quality Evaluation of Chinese Patent Medicine (Traditional Chinese Patent Medicine), Chengdu 611731, China
| | - Huixia Zhang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, National Key Laboratory of Veterinary Public Health Security, Beijing 100193, China
| | - Jingguang Li
- NHC Key Lab of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment (CFSA), Beijing 100022, China
| | - Liu Yang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, National Key Laboratory of Veterinary Public Health Security, Beijing 100193, China
| | - Qiqi Wang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, National Key Laboratory of Veterinary Public Health Security, Beijing 100193, China
| | - Shuai Zhang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, National Key Laboratory of Veterinary Public Health Security, Beijing 100193, China
| | - Pengjie Luo
- NHC Key Lab of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment (CFSA), Beijing 100022, China
| | - Hongping Wang
- Sichuan Institute for Drug Control (Sichuan Testing Center of Medical Devices), NMAP Key Laboratory of Quality Evaluation of Chinese Patent Medicine (Traditional Chinese Patent Medicine), Chengdu 611731, China
| | - Haiyang Jiang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, National Key Laboratory of Veterinary Public Health Security, Beijing 100193, China.
| |
Collapse
|
2
|
Wu M, Chen C, Zhou X, Liu H, Ren Y, Gu J, Lv X, Chen C. Development of disease diagnosis technology based on coattention cross-fusion of multiomics data. Anal Chim Acta 2025; 1351:343919. [PMID: 40187884 DOI: 10.1016/j.aca.2025.343919] [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: 10/26/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Early diagnosis is vital for increasing the rates of curing diseases and patient survival in medicine. With the advancement of biotechnology, the types of bioomics data are increasing. The integration of multiomics data can provide more comprehensive biological information, thereby achieving more accurate diagnoses than single-omics data can. Nevertheless, current multiomics research is often limited to the intelligent diagnosis of a single disease or a few types of omics data and lacks a multiomics disease diagnosis model that can be widely applied to different diseases. Therefore, developing a model that can effectively utilize multiomics data and accurately diagnose diseases has become an important challenge in medical research. RESULTS On the basis of vibrational spectroscopy and metabolomics data, this study proposes an innovative coattention cross-fusion model for disease diagnosis on the basis of interactions of multiomics data. The model not only integrates the information of different omics data but also simulates the interactions between these data to achieve accurate diagnosis of diseases. Through comprehensive experiments, our method achieved accuracies of 95.00 %, 94.95 %, and 97.22 % and area under the curve (AUC) values of 95.00 %, 96.77 %, and 99.31 % on the cervical lymph node metastasis of the thyroid, systemic lupus erythematosus, and cancer datasets, respectively, indicating excellent performance in the diagnosis of multiple diseases. SIGNIFICANCE The proposed model outperforms existing multiomics models, enhancing medical diagnostic accuracy and offering new approaches for multiomics data use in disease diagnosis. The innovative coattention cross-fusion module enables more effective multiomics data processing and analysis, serving as a potent tool for early and precise disease diagnosis with substantial clinical and research implications.
Collapse
Affiliation(s)
- Mingtao Wu
- School of Computer Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Chen Chen
- School of Software, Xinjiang University, Urumqi, 830046, China; Key Laboratory of Signal Detection and Processing, Xinjiang University, Urumqi, 830046, China; Xinjiang Cloud Computing Application Laboratory, Karamay, 834099, China
| | - Xuguang Zhou
- School of Software, Xinjiang University, Urumqi, 830046, China
| | - Hao Liu
- School of Computer Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Yujia Ren
- School of Computer Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Institute for Precision Medicine & Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Xiaoyi Lv
- School of Computer Science and Technology, Xinjiang University, Urumqi, 830046, China; School of Software, Xinjiang University, Urumqi, 830046, China.
| | - Cheng Chen
- School of Software, Xinjiang University, Urumqi, 830046, China.
| |
Collapse
|
3
|
England-Mason G, MacEachern SJ, Amador K, Soomro MH, Reardon AJF, MacDonald AM, Kinniburgh DW, Letourneau N, Giesbrecht GF, Martin JW, Forkert ND, Dewey D. Using machine learning to investigate the influence of the prenatal chemical exposome on neurodevelopment of young children. Neurotoxicology 2025; 108:218-230. [PMID: 40222479 DOI: 10.1016/j.neuro.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
Research investigating the prenatal chemical exposome and child neurodevelopment has typically focused on a limited number of chemical exposures and controlled for sociodemographic factors and maternal mental health. Emerging machine learning approaches may facilitate more comprehensive examinations of the contributions of chemical exposures, sociodemographic factors, and maternal mental health to child neurodevelopment. A machine learning pipeline that utilized feature selection and ranking was applied to investigate which common prenatal chemical exposures and sociodemographic factors best predict neurodevelopmental outcomes in young children. Data from 406 maternal-child pairs enrolled in the APrON study were used. Maternal concentrations of 32 environmental chemical exposures (i.e., phthalates, bisphenols, per- and polyfluoroalkyl substances (PFAS), metals, trace elements) measured during pregnancy and 11 sociodemographic factors, as well as measures of maternal mental health and urinary creatinine were entered into the machine learning pipeline. The pipeline, which consisted of a RReliefF variable selection algorithm and support vector machine regression model, was used to identify and rank the best subset of variables predictive of cognitive, language, and motor development outcomes on the Bayley Scales of Infant Development-Third Edition (Bayley-III) at 2 years of age. Bayley-III cognitive scores were best predicted using 29 variables, resulting in a correlation coefficient of r = 0.27 (R2=0.07). For language outcomes, 45 variables led to the best result (r = 0.30; R2=0.09), whereas for motor outcomes 33 variables led to the best result (r = 0.28, R2=0.09). Environmental chemicals, sociodemographic factors, and maternal mental health were found to be highly ranked predictors of cognitive, language, and motor development in young children. Our findings demonstrate the potential of machine learning approaches to identify and determine the relative importance of different predictors of child neurodevelopmental outcomes. Future developmental neurotoxicology research should consider the prenatal chemical exposome as well as sample characteristics such as sociodemographic factors and maternal mental health as important predictors of child neurodevelopment.
Collapse
Affiliation(s)
- Gillian England-Mason
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sarah J MacEachern
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Psychology, Faculty of Arts, University of Calgary, Calgary, Alberta
| | - Kimberly Amador
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada; Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Munawar Hussain Soomro
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Anthony J F Reardon
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Amy M MacDonald
- Alberta Centre for Toxicology, University of Calgary, Calgary, Alberta, Canada
| | - David W Kinniburgh
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada; Alberta Centre for Toxicology, University of Calgary, Calgary, Alberta, Canada
| | - Nicole Letourneau
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Faculty of Nursing, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, Cumming School of Medicine University of Calgary, Calgary, Alberta, Canada; Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gerald F Giesbrecht
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Psychology, Faculty of Arts, University of Calgary, Calgary, Alberta; Department of Community Health Sciences, Cumming School of Medicine University of Calgary, Calgary, Alberta, Canada
| | - Jonathan W Martin
- Science for Life Laboratory, Department of Environmental Science, Stockholm University, Stockholm, Sweden
| | - Nils D Forkert
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Deborah Dewey
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Community Health Sciences, Cumming School of Medicine University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
4
|
Izuka S, Komai T, Itamiya T, Ota M, Yamada S, Nagafuchi Y, Shoda H, Matsuki K, Yamamoto K, Okamura T, Fujio K. Integration of transcriptome and immunophenotyping data highlights differences in the pathogenetic kinetics of B cells across immune-mediated disease. RMD Open 2025; 11:e005310. [PMID: 40210259 PMCID: PMC11987131 DOI: 10.1136/rmdopen-2024-005310] [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: 11/30/2024] [Accepted: 03/25/2025] [Indexed: 04/12/2025] Open
Abstract
OBJECTIVE To elucidate crucial immune cell subsets and associated immunological pathways by stratifying patients with immune-mediated diseases (IMDs) using immunophenotyping and transcriptomic approaches. METHODS We conducted flow cytometric and transcriptomic analyses in 23 immune cell subsets derived from 235 patients with six IMDs, using our database, utilizing our database, ImmuNexUT. Patients were stratified based on immunophenotyping data. Subsequently, we examined clinical and transcriptomic differences among these stratified clusters. RESULTS Patients with IMDs were stratified into two clusters based on their immunophenotypes. Cluster 1 was enriched with differentiated B cells, including unswitched memory B cells (USM B), switched memory B cells, double-negative B cells and plasmablasts, while cluster 2 was enriched with naïve B cells. Higher disease activity in rheumatoid arthritis and decreased respiratory functions in systemic sclerosis were observed in cluster 1, whereas the disease activity of systemic lupus erythematosus was higher in cluster 2. Numerous differentially expressed genes were detected in USM B. Cluster 1 was associated with glycosylation processes in USM B and elevated B cell-activating factor signalling from myeloid cells in B cells, while cluster 2 exhibited higher B-cell receptor signalling in USM B. Patients in cluster 2, which had an elevated age-associated B-cell signature, exhibited more frequent flares, suggesting that an increased proportion of naïve B cells with this signature is associated with poor prognosis. CONCLUSION Immunophenotyping-based clusters and transcriptome-based states revealed quantitative and qualitative differences in B cells. To predict IMD prognosis, assessing both the quantity and quality of naïve B cells may be crucial.
Collapse
Affiliation(s)
- Shinji Izuka
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshihiko Komai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahiro Itamiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mineto Ota
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Saeko Yamada
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kosuke Matsuki
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Kazuhiko Yamamoto
- Center for Integrative Medical Sciences, the Institute of Physical and Chemical Research (RIKEN), Yokohama, Kanagawa, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Lei Y, Tsang JS. Systems Human Immunology and AI: Immune Setpoint and Immune Health. Annu Rev Immunol 2025; 43:693-722. [PMID: 40279304 DOI: 10.1146/annurev-immunol-090122-042631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
The immune system, critical for human health and implicated in many diseases, defends against pathogens, monitors physiological stress, and maintains tissue and organismal homeostasis. It exhibits substantial variability both within and across individuals and populations. Recent technological and conceptual progress in systems human immunology has provided predictive insights that link personal immune states to intervention responses and disease susceptibilities. Artificial intelligence (AI), particularly machine learning (ML), has emerged as a powerful tool for analyzing complex immune data sets, revealing hidden patterns across biological scales, and enabling predictive models for individualistic immune responses and potentially personalized interventions. This review highlights recent advances in deciphering human immune variation and predicting outcomes, particularly through the concepts of immune setpoint, immune health, and use of the immune system as a window for measuring health. We also provide a brief history of AI; review ML modeling approaches, including their applications in systems human immunology; and explore the potential of AI to develop predictive models and personal immune state embeddings to detect early signs of disease, forecast responses to interventions, and guide personalized health strategies.
Collapse
Affiliation(s)
- Yona Lei
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - John S Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Chan Zuckerberg Biohub NY, New Haven, Connecticut, USA
| |
Collapse
|
6
|
Saegusa K, Tsuchida Y, Komai T, Tsuchiya H, Fujio K. Advances in Targeted Therapy for Systemic Lupus Erythematosus: Current Treatments and Novel Approaches. Int J Mol Sci 2025; 26:929. [PMID: 39940698 PMCID: PMC11816971 DOI: 10.3390/ijms26030929] [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: 12/28/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with diverse clinical manifestations that can lead to severe organ damage. The complex pathophysiology of SLE makes treatment selection difficult. This review examines the current evidence for biological therapies in SLE, including the anti-B cell activating factor antibody belimumab; the type I interferon receptor antagonist anifrolumab; the novel calcineurin inhibitor voclosporin; and rituximab, which targets CD20 on B cells. We also describe emerging therapies, including novel agents in development and CD19-directed chimeric antigen receptor (CAR) T cell therapy, which has shown promise in early clinical experience. Recent advances in biomarker research, including interferon signatures and transcriptomic profiles, may facilitate patient stratification and treatment selection. This review offers insights into current and future treatment strategies for patients with SLE by analyzing clinical trial results and recent immunological findings.
Collapse
Affiliation(s)
| | - Yumi Tsuchida
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; (K.S.); (T.K.); (H.T.); (K.F.)
| | | | | | | |
Collapse
|
7
|
Radziszewska A, Peckham H, Restuadi R, Kartawinata M, Moulding D, de Gruijter NM, Robinson GA, Butt M, Deakin CT, Wilkinson MGL, Wedderburn LR, Jury EC, Rosser EC, Ciurtin C. Type I interferon and mitochondrial dysfunction are associated with dysregulated cytotoxic CD8+ T cell responses in juvenile systemic lupus erythematosus. Clin Exp Immunol 2025; 219:uxae127. [PMID: 39719886 PMCID: PMC11748002 DOI: 10.1093/cei/uxae127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/03/2024] [Accepted: 12/22/2024] [Indexed: 12/26/2024] Open
Abstract
Juvenile systemic lupus erythematosus (JSLE) is an autoimmune condition which causes significant morbidity in children and young adults and is more severe in its presentation than adult-onset SLE. While many aspects of immune dysfunction have been studied extensively in adult-onset SLE, there is limited and contradictory evidence of how cytotoxic CD8+ T cells contribute to disease pathogenesis and studies exploring cytotoxicity in JSLE are virtually non-existent. Here, we report that CD8+ T cell cytotoxic capacity is reduced in JSLE versus healthy controls, irrespective of treatment or disease activity. Transcriptomic and serum metabolomic analysis identified that this reduction in cytotoxic CD8+ T cells in JSLE was associated with upregulated type I interferon (IFN) signalling, mitochondrial dysfunction, and metabolic disturbances when compared to controls. Greater interrogation of the influence of these pathways on altered cytotoxic CD8+ T cell function demonstrated that JSLE CD8+ T cells had enlarged mitochondria and enhanced sensitivity to IFN-α leading to selective apoptosis of effector memory (EM) CD8+ T cells, which are enriched for cytotoxic mediator-expressing cells. This process ultimately contributes to the observed reduction in CD8+ T cell cytotoxicity in JSLE, reinforcing the growing evidence that mitochondrial dysfunction is a key pathogenic factor affecting multiple immune cell populations in type I IFN-driven rheumatic diseases.
Collapse
Affiliation(s)
- Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Restuadi Restuadi
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Melissa Kartawinata
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Dale Moulding
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
- Developmental Biology and Cancer Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nina M de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - George A Robinson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Maryam Butt
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Claire T Deakin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Meredyth G Ll Wilkinson
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Lucy R Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Infection, Immunity and Inflammation Research & Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK
- NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK
| | - Elizabeth C Jury
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
- Department of Ageing, Rheumatology & Regenerative Medicine, Division of Medicine, UCL, London, UK
| |
Collapse
|
8
|
Barlianto W, Wulandari D, Sari TL, Rachmaningrum RR, Asasain RI. Effects of Nigella sativa on disease activity, T lymphocytes and inflammatory cytokine profiles in pediatric systemic lupus erythematosus: A randomized controlled trial. NARRA J 2024; 4:e1063. [PMID: 39816053 PMCID: PMC11731933 DOI: 10.52225/narra.v4i3.1063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/01/2024] [Indexed: 01/30/2025]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with diverse manifestations, requiring long-term treatment that can have side effects, particularly in pediatric patients. Nigella sativa has shown potential for improving SLE symptoms due to its anti-inflammatory and immunomodulatory effects. The aim of this study was to investigate the immunomodulatory effect of N. sativa oil (NSO) on disease activity, T lymphocyte activity and inflammatory cytokine profiles in pediatric SLE patients. A randomized, double-blinded, placebo-controlled clinical trial was conducted at Saiful Anwar Hospital in Malang, Indonesia. Pediatric patients with SLE were randomly assigned to receive either one gram of NSO or a placebo containing starch in capsule form as adjunct therapy alongside their SLE primary treatment. Blood samples were collected before treatment and after eight weeks of daily capsules. Disease activity was assessed using the SLE Disease Activity Index 2000 (SLEDAI-2 K); flow cytometry was used to identify T helper lymphocytes, and serum cytokine levels were measured using ELISA. The statistical analysis tests were performed to compare the outcomes between groups at baseline or after the treatment, and within-group comparisons before and after the study period, as appropriate. A total of 32 patients were included in the study. A significant decrease in the SLEDAI-2 K score was observed at post-treatment in both the NSO and placebo groups (p < 0.001 and p = 0.025, respectively). The percentage of T helper 17 (Th17) cells was significantly reduced in both the NSO and placebo groups post-treatment compared to pre-treatment (p = 0.026 and p = 0.034, respectively). Conversely, the post- treatment percentage of regulatory T (Treg) cells increased significantly in both groups. A significant reduction in interleukin (IL)-2 levels was observed in the NSO and placebo groups at post-treatment compared to pre-treatment (p = 0.006 and p = 0.046, respectively). Additionally, there were increases in IL-4 and IL-6 serum levels in both groups at post-treatment compared to pre-treatment (p < 0.05). This study highlights that although disease activity was not significantly different between NSO and placebo groups, NSO could affect the inflammatory cytokine profiles in pediatric SLE patients.
Collapse
Affiliation(s)
- Wisnu Barlianto
- Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Desy Wulandari
- Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Tita L. Sari
- Department of Pediatrics, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | | | - Rayi I. Asasain
- Department of Pediatrics, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| |
Collapse
|
9
|
Chen C, Wu M, Zuo E, Wu X, Wu L, Liu H, Zhou X, Du Y, Lv X, Chen C. Diagnosis of systemic lupus erythematosus using cross-modal specific transfer fusion technology based on infrared spectra and metabolomics. Anal Chim Acta 2024; 1330:343302. [PMID: 39489981 DOI: 10.1016/j.aca.2024.343302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a chronic autoimmune disease. Currently, the medical diagnosis of SLE mainly relies on the clinical experience of physicians, and there is no universally accepted objective method for diagnosing SLE. Therefore, there is an urgent need to design an intelligent approach to accurately diagnose SLE to assist physicians in formulating appropriate treatment plans. With the rapid development of intelligent medical diagnostic technology, medical data is becoming increasingly multimodal. Multimodal data fusion can provide richer information than single-modal data, and the fusion of multiple modalities can effectively enhance the richness of data features to improve modeling performance. RESULTS In this paper, a cross-modal specific transfer fusion technique based on infrared spectra and metabolomics is proposed to effectively integrate infrared spectra and metabolomics by fully exploiting the intrinsic relationships between features across different modalities, thus achieving the diagnosis of SLE. In this research, a Decision Level Fusion module is also proposed to fuse the representations of two specific transfers further, obtaining the final prediction scores. Comprehensive experimental results demonstrate that the proposed method significantly improves the performance of SLE prediction, with accuracy and Area Under Curve (AUC) reaching 94.98 % and 97.13 %, respectively, outperforming existing methods. SIGNIFICANCE Our framework effectively integrates infrared spectra and metabolomics to achieve a more accurate prediction of SLE. Our research indicates that prediction methods based on different modalities outperform those using single-modality data. The Cross-modal Specific Transfer Fusion module effectively captures the complex relationships within each single modality and models the complex relationships between different modalities.
Collapse
Affiliation(s)
- Cheng Chen
- School of Software, Xinjiang University, Urumqi, 830046, China; People's Hospital of Xinjiang Uyghur Autonomous Region, Xinjiang, China; Xinjiang Key Laboratory of Cardiovascular Homeostasis and Regeneration Research, Xinjiang, China
| | - Mingtao Wu
- School of Computer Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Enguang Zuo
- School of Intelligence Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Xue Wu
- Department of Rheumatology and Immunology, People's Hospital of Xinjiang Uygur Autono-mous Region, Urumqi, Xinjiang, China; Xinjiang Clinical Research Center for Rheumatoid arthritis, Urumqi, Xinjiang, China; Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Lijun Wu
- Department of Rheumatology and Immunology, People's Hospital of Xinjiang Uygur Autono-mous Region, Urumqi, Xinjiang, China; Xinjiang Clinical Research Center for Rheumatoid arthritis, Urumqi, Xinjiang, China
| | - Hao Liu
- School of Software, Xinjiang University, Urumqi, 830046, China
| | - Xuguang Zhou
- School of Software, Xinjiang University, Urumqi, 830046, China
| | - Yang Du
- School of Software, Xinjiang University, Urumqi, 830046, China
| | - Xiaoyi Lv
- School of Software, Xinjiang University, Urumqi, 830046, China; Key Laboratory of signal detection and processing, Xinjiang University, Urumqi, 830046, China
| | - Chen Chen
- School of Software, Xinjiang University, Urumqi, 830046, China.
| |
Collapse
|
10
|
Hammam N, Gheita TA, Bakhiet A, Mahmoud MB, Owaidy RE, Nabi HA, Elsaman AM, Khalifa I, ElBaky AMNEA, Ismail F, Hassan E, El Shereef RR, El-Gazzar II, Moshrif A, Khalil NM, Amer MA, Fathy HM, Salam NA, Elazeem MIA, Hammam O, Fathi HM, Tharwat S. Identifying distinct phenotypes of patients with juvenile systemic lupus erythematosus: results from a cluster analysis by the Egyptian college of rheumatology (ECR) study group. BMC Pediatr 2024; 24:679. [PMID: 39456013 PMCID: PMC11515332 DOI: 10.1186/s12887-024-05137-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
PURPOSE Juvenile systemic lupus erythematosus (J-SLE) is a complex, heterogeneous disease affecting multiple organs. However, the classification of its subgroups is still debated. Therefore, we investigated the aggregated clinical features in patients with J-SLE using cluster analysis. METHODS Patients (≤ 16 years) diagnosed using the Systemic Lupus International Collaborating Clinics (SLICC) classification criteria were identified from the clinical database of the Egyptian College of Rheumatology (ECR) SLE study group. Demographic data, clinical characteristics, laboratory features, and current therapies were selected. A cluster analysis was performed to identify different clinical phenotypes. RESULTS Overall, 404 patients, of whom 355 (87.9%) were female, had a mean age at diagnosis of 11.2 years and a mean disease duration of 2.3 years. We identified four distinct subsets of patients. Patients in cluster 1 (n = 103, 25.5%) were characterized predominantly by mucocutaneous and neurologic manifestations. Patients in cluster 2 (n = 101, 25%) were more likely to have arthritis and pulmonary manifestations. Cluster 3 (n = 71, 17.6%) had the lowest prevalence of arthritis and lupus nephritis (LN), indicative of mild disease intensity. Patients in cluster 4 (n = 129, 31.9%) have the highest frequency of arthritis, vasculitis, and LN. Cluster 1 and 4 patients had the highest disease activity index score and were less likely to use low-dose aspirin (LDA). The SLE damage index was comparable across clusters. CONCLUSIONS Four identified J-SLE clusters express distinct clinical phenotypes. Attention should be paid to including LDA in the therapeutic regimen for J-SLE. Further work is needed to replicate and clarify the phenotype patterns in J-SLE.
Collapse
Affiliation(s)
- Nevin Hammam
- Rheumatology Department, Faculty of Medicine, Assuit University, Assuit, Egypt.
| | - Tamer A Gheita
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ali Bakhiet
- Computer Science Department, Higher Institute of Computer Science and Information Systems, Culture and Science City, Giza, Egypt
| | - Mohamed Bakry Mahmoud
- Computer Science Department, Higher Institute of Computer Science and Information Systems, Culture and Science City, Giza, Egypt
| | - Rasha El Owaidy
- Pediatric Allergy, Immunology and Rheumatology Unit, Children's Hospital, Ain Shams University, Cairo, Egypt
| | - Hend Abdel Nabi
- Pediatrics Department, Rheumatology and Nephrology Unit, Tanta University, Gharbia, Egypt
| | - Ahmed M Elsaman
- Rheumatology Department, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Iman Khalifa
- Pediatrics Unit, Helwan University, Cairo, Egypt
| | - Abeer M Nour ElDin Abd ElBaky
- Pediatrics Department, Medical Research and Clinical Studies Institute, National Research Centre (NRC), Cairo, Egypt
| | - Faten Ismail
- Rheumatology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Eman Hassan
- Internal Medicine Department, Rheumatology Unit, Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Rheumatology Department, Faculty of Medicine, Al-Azhar University, Assuit, Egypt
| | - Rawhya R El Shereef
- Rheumatology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Iman I El-Gazzar
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Abdelhfeez Moshrif
- Internal Medicine Department, Rheumatology Unit, Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Rheumatology Department, Faculty of Medicine, Al-Azhar University, Assuit, Egypt
| | - Noha M Khalil
- Internal Medicine Department, Rheumatology Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Marwa A Amer
- Rheumatology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Hanan M Fathy
- Pediatrics Nephrology Unit, Alexandria University, Alexandria, Egypt
| | - Nancy Abdel Salam
- Pediatrics Nephrology Unit, Alexandria University, Alexandria, Egypt
| | - Mervat I Abd Elazeem
- Rheumatology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Osman Hammam
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, New Valley University, New Valley, Egypt
| | - Hanan M Fathi
- Rheumatology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Samar Tharwat
- Internal Medicine, Rheumatology Unit, Mansoura University, Dakahlia, Egypt
| |
Collapse
|
11
|
Dasgupta S, Dasgupta J. Development of a multivariate predictive nomogram among women with antepartum fetal death diagnosed at ≥ 34 weeks of gestation for outcome of TOLAC. Arch Gynecol Obstet 2024; 310:1131-1139. [PMID: 37930360 DOI: 10.1007/s00404-023-07264-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
OBJECTIVE The present study was planned to develop a nomogram that will give a priori estimate on the probability of vaginal birth from maternal features in women with antepartum fetal death diagnosed at ≥ 34 week's gestation and previous one low transverse cesarean section (LTCS). This will help to reduce maternal complications and increase confidence when planning a trial of labor after cesarean section (TOLAC). METHODS A prospective observational study was planned where participants underwent induction of labor with Foley's catheter (unless already in spontaneous labor) within 24 h of enrolment. Participants with absent or inadequate contractions, oxytocin infusion as an additional agent was used. Data was collected on maternal predelivery features. Outcome of participants was categorized into two classes-vaginal and cesarean delivery. Classifiers were trained with data on maternal features and the accuracy of predicting outcome class determined. The classifier with maximum accuracy was used to develop a nomogram. RESULT Three hundred and one women underwent treatment as per protocol. Two hundred and twenty women attained successful vaginal delivery and eighty-one women underwent caesarean section. Factors having a significant impact on outcome were maternal body mass index (BMI), bishop score, duration of augmentation, estimated foetal weight, interval from previous LTCS, admission to active labor interval, vaginal delivery after LTCS and gestational age. The Naïve -Bayes model gave the highest prediction accuracy (0.88). CONCLUSION Non-linear classifiers by using selective features could predict the outcome of TOLAC among women with intra-uterine fetal death attempting vaginal birth at or beyond 34 weeks gestation with high accuracy.
Collapse
Affiliation(s)
- Subhankar Dasgupta
- Department of Obstetrics and Gynecology, Rampurhat government medical college, New hospital road, Rampurhat, Birbhum, West Bengal, 731224, India.
- Department of Obstetrics and Gynecology, Chittaranjan Seva Sadan, College of Obstetrics, Gynecology and Child Health, Kolkata, India.
- Department of Obstetrics and Gynecology, Medical College, 88, College Street, College square, Kolkata, West Bengal, India, 700073.
| | - Jija Dasgupta
- Department of Obstetrics and Gynecology, Rampurhat government medical college, New hospital road, Rampurhat, Birbhum, West Bengal, 731224, India
- Department of Obstetrics and Gynecology, Chittaranjan Seva Sadan, College of Obstetrics, Gynecology and Child Health, Kolkata, India
- AILABS Adani Enterprises LTD, Kolkata, West Bengal, India
| |
Collapse
|
12
|
Chen YC, Yu HH, Hu YC, Yang YH, Lin YT, Wang LC, Chiang BL, Lee JH. Peripheral blood cells RNA-seq identifies differentially expressed gene network linked to lymphocyte subsets alterations and active lupus nephritis associated with declines in renal function. Heliyon 2024; 10:e32303. [PMID: 38912505 PMCID: PMC11190669 DOI: 10.1016/j.heliyon.2024.e32303] [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: 01/24/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/25/2024] Open
Abstract
Background The aim of this study was to investigate whether quantitative changes in lymphocyte subsets and gene expression in peripheral blood (PB) cells are related to the clinical manifestations and pathogenesis of lupus nephritis (LN). Methods We enrolled 95 pediatric-onset SLE patients with renal involvement who presented with 450 clinical episodes suspicious for LN flare. Percentages of lymphocyte subsets at each episode were determined. We stratified 55 of 95 patients as high or low subset group according to the median percentage of each lymphocyte subset and the association with changes in the eGFR (ΔeGFR) were analyzed. Peripheral blood bulk RNA-seq to identify differentially expressed genes (DEGs) in 9 active LN vs. 9 inactive LN patients and the DEG-derived network was constructed by Ingenuity Pathway Analysis (IPA). Results The mean ΔeGFR of low NK-low memory CD4+ T-high naive CD4+ T group (31.01 mL/min/1.73 m2) was significantly greater than that of high NK-high memory CD4+ T-low naive CD4+ T group (11.83 mL/min/1.73 m2; P = 0.0175). Kaplan-Meier analysis showed that the median time for ΔeGFR decline to mean ΔeGFR is approximately 10 years for high NK-high memory CD4+ T-low naive CD4+ T group and approximately 5 years for low NK-low memory CD4+ T-high naive CD4+ T group (log-rank test P = 0.0294). Conclusions Our study highlighted important connections between DEG-derived network, lymphocyte subset composition, and disease status of LN and GN. A novel scoring system based on lymphocyte subset proportions effectively stratified patients into groups with differential risks for declining renal function.
Collapse
Affiliation(s)
- Yi-Chen Chen
- Fu Jen Catholic University Hospital, New Taipei City, Taiwan, China
| | - Hsin-Hui Yu
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, China
| | - Ya-Chiao Hu
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, China
| | - Yao-Hsu Yang
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, China
| | - Yu-Tsan Lin
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, China
| | - Li-Chieh Wang
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, China
| | - Bor-Luen Chiang
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, China
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan, China
| | - Jyh-Hong Lee
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan, China
| |
Collapse
|
13
|
Khan O, Ajadi JO, Hossain MP. Predicting malaria outbreak in The Gambia using machine learning techniques. PLoS One 2024; 19:e0299386. [PMID: 38753678 PMCID: PMC11098333 DOI: 10.1371/journal.pone.0299386] [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: 08/17/2023] [Accepted: 02/09/2024] [Indexed: 05/18/2024] Open
Abstract
Malaria is the most common cause of death among the parasitic diseases. Malaria continues to pose a growing threat to the public health and economic growth of nations in the tropical and subtropical parts of the world. This study aims to address this challenge by developing a predictive model for malaria outbreaks in each district of The Gambia, leveraging historical meteorological data. To achieve this objective, we employ and compare the performance of eight machine learning algorithms, including C5.0 decision trees, artificial neural networks, k-nearest neighbors, support vector machines with linear and radial kernels, logistic regression, extreme gradient boosting, and random forests. The models are evaluated using 10-fold cross-validation during the training phase, repeated five times to ensure robust validation. Our findings reveal that extreme gradient boosting and decision trees exhibit the highest prediction accuracy on the testing set, achieving 93.3% accuracy, followed closely by random forests with 91.5% accuracy. In contrast, the support vector machine with a linear kernel performs less favorably, showing a prediction accuracy of 84.8% and underperforming in specificity analysis. Notably, the integration of both climatic and non-climatic features proves to be a crucial factor in accurately predicting malaria outbreaks in The Gambia.
Collapse
Affiliation(s)
- Ousman Khan
- Department of Mathematics, College of Computing and Mathematics, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Jimoh Olawale Ajadi
- Department of Mathematics, College of Computing and Mathematics, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
- Interdisciplinary Research Center for Refining & Advanced Chemicals, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - M. Pear Hossain
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| |
Collapse
|
14
|
Horisberger A, Griffith A, Keegan J, Arazi A, Pulford J, Murzin E, Howard K, Hancock B, Fava A, Sasaki T, Ghosh T, Inamo J, Beuschel R, Cao Y, Preisinger K, Gutierrez-Arcelus M, Eisenhaure TM, Guthridge J, Hoover PJ, Dall'Era M, Wofsy D, Kamen DL, Kalunian KC, Furie R, Belmont M, Izmirly P, Clancy R, Hildeman D, Woodle ES, Apruzzese W, McMahon MA, Grossman J, Barnas JL, Payan-Schober F, Ishimori M, Weisman M, Kretzler M, Berthier CC, Hodgin JB, Demeke DS, Putterman C, Brenner MB, Anolik JH, Raychaudhuri S, Hacohen N, James JA, Davidson A, Petri MA, Buyon JP, Diamond B, Zhang F, Lederer JA, Rao DA. Blood immunophenotyping identifies distinct kidney histopathology and outcomes in patients with lupus nephritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.14.575609. [PMID: 38293222 PMCID: PMC10827101 DOI: 10.1101/2024.01.14.575609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Lupus nephritis (LN) is a frequent manifestation of systemic lupus erythematosus, and fewer than half of patients achieve complete renal response with standard immunosuppressants. Identifying non-invasive, blood-based pathologic immune alterations associated with renal injury could aid therapeutic decisions. Here, we used mass cytometry immunophenotyping of peripheral blood mononuclear cells in 145 patients with biopsy-proven LN and 40 healthy controls to evaluate the heterogeneity of immune activation in patients with LN and to identify correlates of renal parameters and treatment response. Unbiased analysis identified 3 immunologically distinct groups of patients with LN that were associated with different patterns of histopathology, renal cell infiltrates, urine proteomic profiles, and treatment response at one year. Patients with enriched circulating granzyme B+ T cells at baseline showed more severe disease and increased numbers of activated CD8 T cells in the kidney, yet they had the highest likelihood of treatment response. A second group characterized primarily by a high type I interferon signature had a lower likelihood of response to therapy, while a third group appeared immunologically inactive by immunophenotyping at enrollment but with chronic renal injuries. Main immune profiles could be distilled down to 5 simple cytometric parameters that recapitulate several of the associations, highlighting the potential for blood immune profiling to translate to clinically useful non-invasive metrics to assess immune-mediated disease in LN.
Collapse
|
15
|
Zhan K, Buhler KA, Chen IY, Fritzler MJ, Choi MY. Systemic lupus in the era of machine learning medicine. Lupus Sci Med 2024; 11:e001140. [PMID: 38443092 PMCID: PMC11146397 DOI: 10.1136/lupus-2023-001140] [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: 12/29/2023] [Accepted: 01/26/2024] [Indexed: 03/07/2024]
Abstract
Artificial intelligence and machine learning applications are emerging as transformative technologies in medicine. With greater access to a diverse range of big datasets, researchers are turning to these powerful techniques for data analysis. Machine learning can reveal patterns and interactions between variables in large and complex datasets more accurately and efficiently than traditional statistical methods. Machine learning approaches open new possibilities for studying SLE, a multifactorial, highly heterogeneous and complex disease. Here, we discuss how machine learning methods are rapidly being integrated into the field of SLE research. Recent reports have focused on building prediction models and/or identifying novel biomarkers using both supervised and unsupervised techniques for understanding disease pathogenesis, early diagnosis and prognosis of disease. In this review, we will provide an overview of machine learning techniques to discuss current gaps, challenges and opportunities for SLE studies. External validation of most prediction models is still needed before clinical adoption. Utilisation of deep learning models, access to alternative sources of health data and increased awareness of the ethics, governance and regulations surrounding the use of artificial intelligence in medicine will help propel this exciting field forward.
Collapse
Affiliation(s)
- Kevin Zhan
- University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Katherine A Buhler
- University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Irene Y Chen
- Computational Precision Health, University of California Berkeley and University of California San Francisco, Berkeley, California, USA
- Electrical Engineering and Computer Science, University of California Berkeley, Berkeley, California, USA
| | - Marvin J Fritzler
- University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - May Y Choi
- University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
- McCaig Institute for Bone and Joint Health, Calgary, Alberta, Canada
| |
Collapse
|
16
|
Xie Q, Li N, Lu Y, Chen J, Qu W, Geng L, Sun L. Prediction of Treatment Effect of SLE-ITP Patients Based on Cost-Sensitive Neural Network and Variational Autoencoder. J Clin Rheumatol 2024:00124743-990000000-00191. [PMID: 38427830 DOI: 10.1097/rhu.0000000000002078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
OBJECTIVE The aim of the study was to examine the factors influencing the therapeutic effect of patients with systemic lupus erythematosus combined with immune thrombocytopenia (SLE-ITP) and develop a prediction model to predict the therapeutic effect of SLE-ITP. METHODS Three hundred twenty-four SLE-ITP patients were retrieved from the electronic health record database of SLE patients in Jiangsu Province according to the latest treatment response criteria for ITP. We adopted the Cox model based on the least absolute shrinkage and selection operator to explore the impact factors affecting patient therapeutic effect, and we developed neural network model to predict therapeutic effect, and in prediction model, cost-sensitivity was introduced to address data category imbalance, and variational autoencoder was used to achieve data augmentation. The performance of each model was evaluated by accuracy and the area under the receiver operator curve. RESULTS The results showed that B-lymphocyte count, H-cholesterol level, complement-3 level, anticardiolipin antibody, and so on could be used as predictors of SLE-ITP curative effect, and abnormal levels of alanine transaminase, immunoglobulin A, and apolipoprotein B predicted adverse treatment response. The neural network treatment effect prediction model based on cost-sensitivity and variational autoencoder was better than the traditional classifiers, with an overall accuracy rate closed to 0.9 and a specificity of more than 0.9, which was useful for clinical practice to identify patients at risk of ineffective treatment response and to achieve better individualized management. CONCLUSIONS By predicting the curative effect of SLE-ITP, the severity of patients can be determined, and then the best treatment strategy can be planned to avoid ineffective treatment.
Collapse
Affiliation(s)
| | - Na Li
- From the Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Youbei Lu
- School of Computer and Information, Hohai University, Nanjing, China
| | - Jiaqi Chen
- School of Computer and Information, Hohai University, Nanjing, China
| | - Wenqiang Qu
- Intelligent Software Research Center, Nanjing Institute of Software Technology, China
| | | | | |
Collapse
|
17
|
Ge Y, Xiao Y, Li M, Yang L, Song P, Li X, Yan H. Maladaptive cognitive regulation moderates the mediating role of emotion dysregulation on the association between psychosocial factors and non-suicidal self-injury in depression. Front Psychiatry 2023; 14:1279108. [PMID: 38098637 PMCID: PMC10719840 DOI: 10.3389/fpsyt.2023.1279108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/10/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction Non-suicidal self-injury (NSSI) is highly prevalent in depression, and is associated with psychosocial factors, emotion dysregulation, and strategies of cognitive emotion regulation. However, the internal combination and interactions of these risk factors in depression remain unclear. Methods Data from 122 patients with depression, including 56 with NSSI and 66 without NSSI, were analyzed. Self-rating scales were used to assess psychosocial factors, emotion dysregulation, and cognitive regulation strategies. Sparse partial least squares discriminant analysis (sPLS-DA) was employed to explore internal combinations in each profile. A moderated mediation model was applied to examine their interactional relationship. Results The results identified an NSSI-related psychosocial profile characterized by high neuroticism, childhood trauma, poor family functioning, and low psychological resilience. Emotion dysregulation, including high levels of alexithymia, anhedonia, and emotion regulation difficulties, mediated the association between this psychosocial profile and NSSI. The mediated effect was further moderated by maladaptive cognitive regulation strategies. Limitations Lack of sufficient information on NSSI frequency and severity. Relatively small sample size for discussing the impact of gender and age of depressive patients with NSSI. Conclusion These findings hold important implications for the prevention, treatment, and rehabilitation of NSSI.
Collapse
Affiliation(s)
- Yuqi Ge
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Yang Xiao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Mingzhu Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Lei Yang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Peihua Song
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Xueni Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Hao Yan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health, National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| |
Collapse
|
18
|
Meloni A, Parravano M, Pistoia L, Cossu A, Grassedonio E, Renne S, Fina P, Spasiano A, Salvo A, Bagnato S, Gerardi C, Borsellino Z, Cademartiri F, Positano V. Phenotypic Clustering of Beta-Thalassemia Intermedia Patients Using Cardiovascular Magnetic Resonance. J Clin Med 2023; 12:6706. [PMID: 37959172 PMCID: PMC10647397 DOI: 10.3390/jcm12216706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 11/15/2023] Open
Abstract
We employed an unsupervised clustering method that integrated demographic, clinical, and cardiac magnetic resonance (CMR) data to identify distinct phenogroups (PGs) of patients with beta-thalassemia intermedia (β-TI). We considered 138 β-TI patients consecutively enrolled in the Myocardial Iron Overload in Thalassemia (MIOT) Network who underwent MR for the quantification of hepatic and cardiac iron overload (T2* technique), the assessment of biventricular size and function and atrial dimensions (cine images), and the detection of replacement myocardial fibrosis (late gadolinium enhancement technique). Three mutually exclusive phenogroups were identified based on unsupervised hierarchical clustering of principal components: PG1, women; PG2, patients with replacement myocardial fibrosis, increased biventricular volumes and masses, and lower left ventricular ejection fraction; and PG3, men without replacement myocardial fibrosis, but with increased biventricular volumes and masses and lower left ventricular ejection fraction. The hematochemical parameters and the hepatic and cardiac iron levels did not contribute to the PG definition. PG2 exhibited a significantly higher risk of future cardiovascular events (heart failure, arrhythmias, and pulmonary hypertension) than PG1 (hazard ratio-HR = 10.5; p = 0.027) and PG3 (HR = 9.0; p = 0.038). Clustering emerged as a useful tool for risk stratification in TI, enabling the identification of three phenogroups with distinct clinical and prognostic characteristics.
Collapse
Affiliation(s)
- Antonella Meloni
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, PI, Italy; (L.P.); (F.C.); (V.P.)
- Unità Operativa Complessa Bioingegneria, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, PI, Italy;
| | - Michela Parravano
- Unità Operativa Complessa Bioingegneria, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, PI, Italy;
- Dipartimento di Ingegneria dell’Informazione, Università degli Studi di Pisa, 56122 Pisa, PI, Italy
| | - Laura Pistoia
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, PI, Italy; (L.P.); (F.C.); (V.P.)
- Unità Operativa Complessa Ricerca Clinica, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, PI, Italy
| | - Alberto Cossu
- Unità Operativa Radiologia Universitaria, Azienda Ospedaliero-Universitaria “S. Anna”, 44124 Cona, FE, Italy;
| | - Emanuele Grassedonio
- Sezione di Scienze Radiologiche, Dipartimento di Biopatologia e Biotecnologie Mediche, Policlinico “Paolo Giaccone”, 90127 Palermo, PA, Italy;
| | - Stefania Renne
- Struttura Complessa di Cardioradiologia-UTIC, Presidio Ospedaliero “Giovanni Paolo II”, 88046 Lamezia Terme, CZ, Italy;
| | - Priscilla Fina
- Unità Operativa Complessa Diagnostica per Immagini, Ospedale “Sandro Pertini”, 00157 Roma, RM, Italy;
| | - Anna Spasiano
- Unità Operativa Semplice Dipartimentale Malattie Rare del Globulo Rosso, Azienda Ospedaliera di Rilievo Nazionale “A. Cardarelli”, 80131 Napoli, NA, Italy;
| | - Alessandra Salvo
- Unità Operativa Semplice Talassemia, Presidio Ospedaliero “Umberto I”, 96100 Siracusa, SR, Italy;
| | - Sergio Bagnato
- Ematologia Microcitemia, Ospedale San Giovanni di Dio—ASP Crotone, 88900 Crotone, KR, Italy;
| | - Calogera Gerardi
- Unità Operativa Semplice Dipartimentale di Talassemia, Presidio Ospedaliero “Giovanni Paolo II”—Distretto AG2 di Sciacca, 92019 Sciacca, AG, Italy;
| | - Zelia Borsellino
- Unità Operativa Complessa Ematologia con Talassemia, ARNAS Civico “Benfratelli-Di Cristina”, 90134 Palermo, PA, Italy;
| | - Filippo Cademartiri
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, PI, Italy; (L.P.); (F.C.); (V.P.)
| | - Vincenzo Positano
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, PI, Italy; (L.P.); (F.C.); (V.P.)
- Unità Operativa Complessa Bioingegneria, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, PI, Italy;
- Dipartimento di Ingegneria dell’Informazione, Università degli Studi di Pisa, 56122 Pisa, PI, Italy
| |
Collapse
|
19
|
Renaudineau Y, Brooks W, Belliere J. Lupus Nephritis Risk Factors and Biomarkers: An Update. Int J Mol Sci 2023; 24:14526. [PMID: 37833974 PMCID: PMC10572905 DOI: 10.3390/ijms241914526] [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: 09/01/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Lupus nephritis (LN) represents the most severe organ manifestation of systemic lupus erythematosus (SLE) in terms of morbidity and mortality. To reduce these risks, tremendous efforts have been made in the last decade to characterize the different steps of the disease and to develop biomarkers in order to better (i) unravel the pre-SLE stage (e.g., anti-nuclear antibodies and interferon signature); (ii) more timely initiation of therapy by improving early and accurate LN diagnosis (e.g., pathologic classification was revised); (iii) monitor disease activity and therapeutic response (e.g., recommendation to re-biopsy, new urinary biomarkers); (iv) prevent disease flares (e.g., serologic and urinary biomarkers); (v) mitigate the deterioration in the renal function; and (vi) reduce side effects with new therapeutic guidelines and novel therapies. However, progress is poor in terms of improvement with early death attributed to active SLE or infections, while later deaths are related to the chronicity of the disease and the use of toxic therapies. Consequently, an individualized treat-to-target strategy is mandatory, and for that, there is an unmet need to develop a set of accurate biomarkers to be used as the standard of care and adapted to each stage of the disease.
Collapse
Affiliation(s)
- Yves Renaudineau
- Department of Immunology, Referral Medical Biology Laboratory, University Hospital of Toulouse, Institut National de la Santé Et de la Recherche Médicale (INSERM) U1291, Centre National de la Recherche Scientifique (CNRS) U5051, 31400 Toulouse, France
| | - Wesley Brooks
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA;
| | - Julie Belliere
- Department of Nephrology and Organ Transplantation, Referral Centre for Rare Kidney Diseases, University Hospital of Toulouse, INSERM U1297, 31400 Toulouse, France;
| |
Collapse
|
20
|
Wang DC, Xu WD, Qin Z, Fu L, Lan YY, Liu XY, Huang AF. Systemic lupus erythematosus with high disease activity identification based on machine learning. Inflamm Res 2023; 72:1909-1918. [PMID: 37725103 DOI: 10.1007/s00011-023-01793-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023] Open
Abstract
OBJECTIVE Clinical evaluation of systemic lupus erythematosus (SLE) disease activity is limited and inconsistent, and high disease activity significantly, seriously impacts on SLE patients. This study aims to generate a machine learning model to identify SLE patients with high disease activity. METHOD A total of 1014 SLE patients with low disease activity and 453 SLE patients with high disease activity were included. A total of 94 clinical, laboratory data and 17 meteorological indicators were collected. After data preprocessing, we use mutual information and multisurf to evaluate and select the importance of features. The selected features are used for machine learning modeling. Performance of the model is evaluated and verified by a series of binary classification indicators. RESULTS We screened out hematuria, proteinuria, pyuria, low complement, precipitation, sunlight and other features for model construction by integrated feature selection. After hyperparameter optimization, the LGB has the best performance (ROC: AUC = 0.930; PRC: AUC = 0.911, APS = 0.913; balance accuracy: 0.856), and the worst is the naive bayes (ROC: AUC = 0.849; PRC: AUC = 0.719, APS = 0.714; balance accuracy: 0.705). Finally, the selection of features has good consistency in the composite feature importance bar plot. CONCLUSION We identify SLE patients with high disease activity by a simple machine learning pipeline, especially the LGB model based on the characteristics of proteinuria, hematuria, pyuria and other feathers screened out by collective feature selection.
Collapse
Affiliation(s)
- Da-Cheng Wang
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, 646000, Sichuan, China
| | - Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, 646000, Sichuan, China.
| | - Zhen Qin
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, Sichuan, China
| | - Lu Fu
- Laboratory Animal Center, Southwest Medical University, 1 Xianglin Road, Luzhou, 646000, Sichuan, China
| | - You-Yu Lan
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, Sichuan, China
| | - Xiao-Yan Liu
- Department of Evidence-Based Medicine, Southwest Medical University, 1 Xianglin Road, Luzhou, 646000, Sichuan, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, 25 Taiping Road, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
21
|
Qiao J, Zhang SX, Chang MJ, Zhao R, Song S, Hao JW, Wang C, Hu JX, Gao C, Wang CH, Li XF. Deep stratification by transcriptome molecular characters for precision treatment of patients with systemic lupus erythematosus. Rheumatology (Oxford) 2023; 62:2574-2584. [PMID: 36308437 DOI: 10.1093/rheumatology/keac625] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/18/2022] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVES To leverage the high clinical heterogeneity of systemic lupus erythematosus (SLE), we developed and validated a new stratification scheme by integrating genome-scale transcriptomic profiles to identify patient subtypes sharing similar transcriptomic markers and drug targets. METHODS A normalized compendium of transcription profiles was created from peripheral blood mononuclear cells (PBMCs) of 1046 SLE patients and 86 healthy controls (HCs), covering an intersection of 13 689 genes from six microarray datasets. Upregulated differentially expressed genes were subjected to functional and network analysis in which samples were grouped using unsupervised clustering to identify patient subtypes. Then, clustering stability was evaluated by the stratification of six integrated RNA-sequencing datasets using the same method. Finally, the Xgboost classifier was applied to the independent datasets to identify factors associated with treatment outcomes. RESULTS Based on 278 upregulated DEGs of the transcript profiles, SLE patients were classified into three subtypes (subtype A-C) each with distinct molecular and cellular signatures. Neutrophil activation-related pathways were markedly activated in subtype A (named NE-driving), whereas lymphocyte and IFN-related pathways were more enriched in subtype B (IFN-driving). As the most severe subtype, subtype C [NE-IFN-dual-driving (Dual-driving)] shared functional mechanisms with both NE-driving and IFN-driving, which was closely associated with clinical features and could be used to predict the responses of treatment. CONCLUSION We developed the largest cohesive SLE transcriptomic compendium for deep stratification using the most comprehensive microarray and RNA sequencing datasets to date. This result could guide future design of molecular diagnosis and the development of stratified therapy for SLE patients.
Collapse
Affiliation(s)
- Jun Qiao
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Min-Jing Chang
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Rong Zhao
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Shan Song
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Jia-Wei Hao
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Can Wang
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Big Data for Clinical Decision, Shanxi Medical University, Taiyuan, China
| | - Jing-Xi Hu
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Cai-Hong Wang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| | - Xiao-Feng Li
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, China
- Ministry of Education, Key Laboratory of Cellular Physiology at Shanxi Medical University, Taiyuan, China
| |
Collapse
|
22
|
Akthar M, Nair N, Carter LM, Vital EM, Sutton E, McHugh N, Bruce IN, Reynolds JA. Deconvolution of whole blood transcriptomics identifies changes in immune cell composition in patients with systemic lupus erythematosus (SLE) treated with mycophenolate mofetil. Arthritis Res Ther 2023; 25:111. [PMID: 37391799 PMCID: PMC10311871 DOI: 10.1186/s13075-023-03089-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/09/2023] [Indexed: 07/02/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a clinically and biologically heterogeneous autoimmune disease. We explored whether the deconvolution of whole blood transcriptomic data could identify differences in predicted immune cell frequency between active SLE patients, and whether these differences are associated with clinical features and/or medication use. METHODS Patients with active SLE (BILAG-2004 Index) enrolled in the BILAG-Biologics Registry (BILAG-BR), prior to change in therapy, were studied as part of the MASTERPLANS Stratified Medicine consortium. Whole blood RNA-sequencing (RNA-seq) was conducted at enrolment into the registry. Data were deconvoluted using CIBERSORTx. Predicted immune cell frequencies were compared between active and inactive disease in the nine BILAG-2004 domains and according to immunosuppressant use (current and past). RESULTS Predicted cell frequency varied between 109 patients. Patients currently, or previously, exposed to mycophenolate mofetil (MMF) had fewer inactivated macrophages (0.435% vs 1.391%, p = 0.001), naïve CD4 T cells (0.961% vs 2.251%, p = 0.002), and regulatory T cells (1.858% vs 3.574%, p = 0.007), as well as a higher proportion of memory activated CD4 T cells (1.826% vs 1.113%, p = 0.015), compared to patients never exposed to MMF. These differences remained statistically significant after adjusting for age, gender, ethnicity, disease duration, renal disease, and corticosteroid use. There were 2607 differentially expressed genes (DEGs) in patients exposed to MMF with over-representation of pathways relating to eosinophil function and erythrocyte development and function. Within CD4 + T cells, there were fewer predicted DEGs related to MMF exposure. No significant differences were observed for the other conventional immunosuppressants nor between patients according disease activity in any of the nine organ domains. CONCLUSION MMF has a significant and persisting effect on the whole blood transcriptomic signature in patients with SLE. This highlights the need to adequately adjust for background medication use in future studies using whole blood transcriptomics.
Collapse
Affiliation(s)
- Mumina Akthar
- Rheumatology Department, Sandwell and West Birmingham NHS Trust, Birmingham, UK
| | - Nisha Nair
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Lucy M Carter
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Edward M Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Emily Sutton
- Centre for Epidemiology Versus Arthritis, Division of Musculoskeletal & Dermatological Sciences, The University of Manchester, Manchester, UK
| | - Neil McHugh
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Ian N Bruce
- Centre for Epidemiology Versus Arthritis, Division of Musculoskeletal & Dermatological Sciences, The University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - John A Reynolds
- Rheumatology Department, Sandwell and West Birmingham NHS Trust, Birmingham, UK.
- Rheumatology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
23
|
Fasano S, Milone A, Nicoletti GF, Isenberg DA, Ciccia F. Precision medicine in systemic lupus erythematosus. Nat Rev Rheumatol 2023; 19:331-342. [PMID: 37041269 DOI: 10.1038/s41584-023-00948-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/13/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that has diverse clinical manifestations, ranging from restricted cutaneous involvement to life-threatening systemic organ involvement. The heterogeneity of pathomechanisms that lead to SLE contributes to between-patient variation in clinical phenotype and treatment response. Ongoing efforts to dissect cellular and molecular heterogeneity in SLE could facilitate the future development of stratified treatment recommendations and precision medicine, which is a considerable challenge for SLE. In particular, some genes involved in the clinical heterogeneity of SLE and some phenotype-related loci (STAT4, IRF5, PDGF genes, HAS2, ITGAM and SLC5A11) have an association with clinical features of the disease. An important part is also played by epigenetic varation (in DNA methylation, histone modifications and microRNAs) that influences gene expression and affects cell function without modifying the genome sequence. Immune profiling can help to identify an individual's specific response to a therapy and can potentially predict outcomes, using techniques such as flow cytometry, mass cytometry, transcriptomics, microarray analysis and single-cell RNA sequencing. Furthermore, the identification of novel serum and urinary biomarkers would enable the stratification of patients according to predictions of long-term outcomes and assessments of potential response to therapy.
Collapse
Affiliation(s)
- Serena Fasano
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| | - Alessandra Milone
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanni Francesco Nicoletti
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - David A Isenberg
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| | - Francesco Ciccia
- Rheumatology Unit, Department of Precision Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
24
|
Wahadat MJ, van Tilburg SJ, Mueller YM, de Wit H, Van Helden-Meeuwsen CG, Langerak AW, Gruijters MJ, Mubarak A, Verkaaik M, Katsikis PD, Versnel MA, Kamphuis S. Targeted multiomics in childhood-onset SLE reveal distinct biological phenotypes associated with disease activity: results from an explorative study. Lupus Sci Med 2023; 10:10/1/e000799. [PMID: 37012057 PMCID: PMC10083882 DOI: 10.1136/lupus-2022-000799] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 02/10/2023] [Indexed: 04/05/2023]
Abstract
OBJECTIVE To combine targeted transcriptomic and proteomic data in an unsupervised hierarchical clustering method to stratify patients with childhood-onset SLE (cSLE) into similar biological phenotypes, and study the immunological cellular landscape that characterises the clusters. METHODS Targeted whole blood gene expression and serum cytokines were determined in patients with cSLE, preselected on disease activity state (at diagnosis, Low Lupus Disease Activity State (LLDAS), flare). Unsupervised hierarchical clustering, agnostic to disease characteristics, was used to identify clusters with distinct biological phenotypes. Disease activity was scored by clinical SELENA-SLEDAI (Safety of Estrogens in Systemic Lupus Erythematosus National Assessment-Systemic Lupus Erythematosus Disease Activity Index). High-dimensional 40-colour flow cytometry was used to identify immune cell subsets. RESULTS Three unique clusters were identified, each characterised by a set of differentially expressed genes and cytokines, and by disease activity state: cluster 1 contained primarily patients in LLDAS, cluster 2 contained mainly treatment-naïve patients at diagnosis and cluster 3 contained a mixed group of patients, namely in LLDAS, at diagnosis and disease flare. The biological phenotypes did not reflect previous organ system involvement and over time, patients could move from one cluster to another. Healthy controls clustered together in cluster 1. Specific immune cell subsets, including CD11c+ B cells, conventional dendritic cells, plasmablasts and early effector CD4+ T cells, differed between the clusters. CONCLUSION Using a targeted multiomic approach, we clustered patients into distinct biological phenotypes that are related to disease activity state but not to organ system involvement. This supports a new concept where choice of treatment and tapering strategies are not solely based on clinical phenotype but includes measuring novel biological parameters.
Collapse
Affiliation(s)
- Mohamed Javad Wahadat
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
- Department of Paediatric Rheumatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | - Yvonne M Mueller
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Harm de Wit
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Anton W Langerak
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Marike J Gruijters
- Department of Paediatric Rheumatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Amani Mubarak
- Department of Paediatric Rheumatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Marleen Verkaaik
- Department of Paediatric Rheumatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Peter D Katsikis
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Marjan A Versnel
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Sylvia Kamphuis
- Department of Paediatric Rheumatology, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
25
|
Visibelli A, Roncaglia B, Spiga O, Santucci A. The Impact of Artificial Intelligence in the Odyssey of Rare Diseases. Biomedicines 2023; 11:887. [PMID: 36979866 PMCID: PMC10045927 DOI: 10.3390/biomedicines11030887] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Emerging machine learning (ML) technologies have the potential to significantly improve the research and treatment of rare diseases, which constitute a vast set of diseases that affect a small proportion of the total population. Artificial Intelligence (AI) algorithms can help to quickly identify patterns and associations that would be difficult or impossible for human analysts to detect. Predictive modeling techniques, such as deep learning, have been used to forecast the progression of rare diseases, enabling the development of more targeted treatments. Moreover, AI has also shown promise in the field of drug development for rare diseases with the identification of subpopulations of patients who may be most likely to respond to a particular drug. This review aims to highlight the achievements of AI algorithms in the study of rare diseases in the past decade and advise researchers on which methods have proven to be most effective. The review will focus on specific rare diseases, as defined by a prevalence rate that does not exceed 1-9/100,000 on Orphanet, and will examine which AI methods have been most successful in their study. We believe this review can guide clinicians and researchers in the successful application of ML in rare diseases.
Collapse
Affiliation(s)
- Anna Visibelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Bianca Roncaglia
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Ottavia Spiga
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Competence Center ARTES 4.0, 53100 Siena, Italy
- SienabioACTIVE—SbA, 53100 Siena, Italy
| | - Annalisa Santucci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Competence Center ARTES 4.0, 53100 Siena, Italy
- SienabioACTIVE—SbA, 53100 Siena, Italy
| |
Collapse
|
26
|
Yaung KN, Yeo JG, Kumar P, Wasser M, Chew M, Ravelli A, Law AHN, Arkachaisri T, Martini A, Pisetsky DS, Albani S. Artificial intelligence and high-dimensional technologies in the theragnosis of systemic lupus erythematosus. THE LANCET. RHEUMATOLOGY 2023; 5:e151-e165. [PMID: 38251610 DOI: 10.1016/s2665-9913(23)00010-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 12/14/2022] [Accepted: 01/04/2023] [Indexed: 02/22/2023]
Abstract
Systemic lupus erythematosus is a complex, systemic autoimmune disease characterised by immune dysregulation. Pathogenesis is multifactorial, contributing to clinical heterogeneity and posing challenges for diagnosis and treatment. Although strides in treatment options have been made in the past 15 years, with the US Food and Drug Administration approval of belimumab in 2011, there are still many patients who have inadequate responses to therapy. A better understanding of underlying disease mechanisms with a holistic and multiparametric approach is required to improve clinical assessment and treatment. This Review discusses the evolution of genomics, epigenomics, transcriptomics, and proteomics in the study of systemic lupus erythematosus and ways to amalgamate these silos of data with a systems-based approach while also discussing ways to strengthen the overall process. These mechanistic insights will facilitate the discovery of functionally relevant biomarkers to guide rational therapeutic selection to improve patient outcomes.
Collapse
Affiliation(s)
- Katherine Nay Yaung
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore; Duke-NUS Medical School, Singapore.
| | - Joo Guan Yeo
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore; Duke-NUS Medical School, Singapore; Rheumatology and Immunology Service, KK Women's and Children's Hospital, Singapore
| | - Pavanish Kumar
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Martin Wasser
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Marvin Chew
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Angelo Ravelli
- Direzione Scientifica, IRCCS Istituto Giannina Gaslini, Genoa, Italy; Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Università degli Studi di Genova, Genoa, Italy
| | - Annie Hui Nee Law
- Duke-NUS Medical School, Singapore; Department of Rheumatology and Immunology, Singapore General Hospital, Singapore
| | - Thaschawee Arkachaisri
- Duke-NUS Medical School, Singapore; Rheumatology and Immunology Service, KK Women's and Children's Hospital, Singapore
| | | | - David S Pisetsky
- Department of Medicine and Department of Immunology, Duke University Medical Center, Durham, NC, USA; Medical Research Service, Veterans Administration Medical Center, Durham, NC, USA
| | - Salvatore Albani
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore; Duke-NUS Medical School, Singapore; Rheumatology and Immunology Service, KK Women's and Children's Hospital, Singapore
| |
Collapse
|
27
|
Munguía-Realpozo P, Etchegaray-Morales I, Mendoza-Pinto C, Méndez-Martínez S, Osorio-Peña ÁD, Ayón-Aguilar J, García-Carrasco M. Current state and completeness of reporting clinical prediction models using machine learning in systemic lupus erythematosus: A systematic review. Autoimmun Rev 2023; 22:103294. [PMID: 36791873 DOI: 10.1016/j.autrev.2023.103294] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Abstract
OBJECTIVE We carried out a systematic review (SR) of adherence in diagnostic and prognostic applications of ML in SLE using the Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis (TRIPOD) Statement. METHODS A SR employing five databases was conducted from its inception until December 2021. We identified articles that evaluated the utilization of ML for prognostic and/or diagnostic purposes. This SR was reported based on the PRISMA guidelines. The TRIPOD statement assessed adherence to reporting standards. Assessment for risk of bias was done using PROBAST tool. RESULTS We included 45 studies: 29 (64.4%) diagnostic and 16 (35.5%) prognostic prediction- model studies. Overall, articles adhered by between 17% and 67% (median 43%, IQR 37-49%) to TRIPOD items. Only few articles reported the model's predictive performance (2.3%, 95% CI 0.06-12.0), testing of interaction terms (2.3%, 95% CI 0.06-12.0), flow of participants (50%, 95% CI; 34.6-65.4), blinding of predictors (2.3%, 95% CI 0.06-12.0), handling of missing data (36.4%, 95% CI 22.4-52.2), and appropriate title (20.5%, 95% CI 9.8-35.3). Some items were almost completely reported: the source of data (88.6%, 95% CI 75.4-96.2), eligibility criteria (86.4%, 95% CI 76.2-96.5), and interpretation of findings (88.6%, 95% CI 75.4-96.2). In addition, most of model studies had high risk of bias. CONCLUSIONS The reporting adherence of ML-based model developed for SLE, is currently inadequate. Several items deemed crucial for transparent reporting were not fully reported in studies on ML-based prediction models. REVIEW REGISTRATION PROSPERO ID# CRD42021284881. (Amended to limit the scope).
Collapse
Affiliation(s)
- Pamela Munguía-Realpozo
- Systemic Autoimmune Diseases Research Unit, Specialties Hospital UMAE- CIBIOR, Mexican Institute for Social Security, Puebla, Mexico; Department of Rheumatology, Medicine School, Meritorious Autonomous University of Puebla, Mexico
| | - Ivet Etchegaray-Morales
- Department of Rheumatology, Medicine School, Meritorious Autonomous University of Puebla, Mexico.
| | - Claudia Mendoza-Pinto
- Systemic Autoimmune Diseases Research Unit, Specialties Hospital UMAE- CIBIOR, Mexican Institute for Social Security, Puebla, Mexico; Department of Rheumatology, Medicine School, Meritorious Autonomous University of Puebla, Mexico.
| | | | - Ángel David Osorio-Peña
- Department of Rheumatology, Medicine School, Meritorious Autonomous University of Puebla, Mexico
| | - Jorge Ayón-Aguilar
- Coordination of Health Research, Mexican Social Security Institute, Puebla, Mexico.
| | - Mario García-Carrasco
- Department of Rheumatology, Medicine School, Meritorious Autonomous University of Puebla, Mexico
| |
Collapse
|
28
|
Nelson AE, Arbeeva L. Narrative Review of Machine Learning in Rheumatic and Musculoskeletal Diseases for Clinicians and Researchers: Biases, Goals, and Future Directions. J Rheumatol 2022; 49:1191-1200. [PMID: 35840150 PMCID: PMC9633365 DOI: 10.3899/jrheum.220326] [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] [Accepted: 06/21/2022] [Indexed: 11/22/2022]
Abstract
There has been rapid growth in the use of artificial intelligence (AI) analytics in medicine in recent years, including in rheumatic and musculoskeletal diseases (RMDs). Such methods represent a challenge to clinicians, patients, and researchers, given the "black box" nature of most algorithms, the unfamiliarity of the terms, and the lack of awareness of potential issues around these analyses. Therefore, this review aims to introduce this subject area in a way that is relevant and meaningful to clinicians and researchers. We hope to provide some insights into relevant strengths and limitations, reporting guidelines, as well as recent examples of such analyses in key areas, with a focus on lessons learned and future directions in diagnosis, phenotyping, prognosis, and precision medicine in RMDs.
Collapse
Affiliation(s)
- Amanda E Nelson
- A.E. Nelson, MD, MSCR, Department of Medicine, Division of Rheumatology, Allergy, and Immunology, University of North Carolina at Chapel Hill;
| | - Liubov Arbeeva
- L. Arbeeva, MS, Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
29
|
Robinson GA, Peng J, Peckham H, Butler G, Pineda-Torra I, Ciurtin C, Jury EC. Investigating sex differences in T regulatory cells from cisgender and transgender healthy individuals and patients with autoimmune inflammatory disease: a cross-sectional study. THE LANCET. RHEUMATOLOGY 2022; 4:e710-e724. [PMID: 36353692 PMCID: PMC9633330 DOI: 10.1016/s2665-9913(22)00198-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Sexual dimorphisms, which vary depending on age group and pubertal status, have been described across both the innate and adaptive immune system. We explored the influence of sex hormones on immune phenotype in the context of adolescent health and autoimmunity. Methods In this cross-sectional study, healthy, post-pubertal cisgender individuals (aged 16-25 years); healthy, pre-pubertal cisgender individuals (aged 6-11 years); transgender individuals (aged 18-19 years) undergoing gender-affirming treatment (testosterone in individuals assigned female sex at birth and oestradiol in individuals assigned male sex at birth); and post-pubertal cisgender individuals (aged 14-25 years) with juvenile-onset systemic lupus erythematosus (SLE) age-matched to cisgender individuals without juvenile-onset SLE were eligible for inclusion. Frequencies of 28 immune-cell subsets (including different T cell, B cell, and monocyte subsets) from each participant were measured in peripheral blood mononuclear cells by flow cytometry and analysed by balanced random forest machine learning. RNA-sequencing was used to compare sex and gender differences in regulatory T (Treg) cell phenotype between participants with juvenile-onset SLE, age-matched cis-gender participants without the disease, and age matched transgender individuals on gender-affirming sex hormone treatment. Differentially expressed genes were analysed by cluster and pathway analysis. Suppression assays assessed the anti-inflammatory function of Treg cells in vitro. Findings Between Sept 5, 2012, and Nov 6, 2019, peripheral blood was collected from 39 individuals in the post-pubertal group (17 [44%] cisgender men, mean age 18·76 years [SD 2·66]; 22 [56%] cisgender women, mean age 18·59 years [2·81]), 14 children in the cisgender pre-pubertal group (seven [50%] cisgender boys, mean age 8·90 [1·66]; seven [50%] cisgender girls, mean age 8·40 [1·58]), ten people in the transgender group (five [50%] transgender men, mean age 18·20 years [0·47]; five [50%] transgender women, mean age 18·70 years [0·55]), and 35 people in the juvenile-onset SLE group (12 [34%] cisgender men, mean age 18·58 years [2·35]; 23 [66%] cisgender women, mean age 19·48 [3·08]). Statistically significantly elevated frequencies of Treg cells were one of the top immune-cell features differentiating young post-pubertal cisgender men from similarly aged cisgender women (p=0·0097). Treg cells from young cisgender men had a statistically significantly increased suppressive capacity in vitro compared with those from cisgender women and a distinct transcriptomic signature significantly enriched for genes in the PI3K-AKT signalling pathway. Gender-affirming sex hormones in transgender men and transgender women induced multiple statistically significant changes in the Treg-cell transcriptome, many of which enriched functional pathways that overlapped with those altered between cisgender men and cisgender women, highlighting a hormonal influence on Treg-cell function by gender. Finally, sex differences in Treg-cell frequency were absent and suppressive capacity was reversed in patients with juvenile-onset SLE, but sex differences in Treg-cell transcriptional signatures were significantly more pronounced in patients with juvenile-onset SLE compared with individuals without juvenile-onset SLE, suggesting that sex hormone signalling could be dysregulated in autoimmunity. Interpretation Sex-chromosomes and hormones might drive changes in Treg-cell frequency and function. Young post-pubertal men have a more anti-inflammatory Treg-cell profile, which could explain inflammatory disease susceptibilities, and inform sex-tailored therapeutic strategies. Funding Versus Arthritis, UK National Institute for Health Research University College London Hospital Biomedical Research Centre, Lupus UK, and The Rosetrees Trust.
Collapse
Affiliation(s)
- George A Robinson
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Junjie Peng
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Hannah Peckham
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Gary Butler
- Department of Paediatric and Adolescent Endocrinology, University College London Hospital and Great Ormond Street Institute of Child Health, University College London, London, UK
- Gender Identity Development Service, Tavistock and Portman NHS Foundation Trust, London, UK
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, Division of Medicine, University College London, London, UK
| | - Coziana Ciurtin
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, UK
| |
Collapse
|
30
|
Radziszewska A, Moulder Z, Jury EC, Ciurtin C. CD8 + T Cell Phenotype and Function in Childhood and Adult-Onset Connective Tissue Disease. Int J Mol Sci 2022; 23:11431. [PMID: 36232733 PMCID: PMC9569696 DOI: 10.3390/ijms231911431] [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: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
CD8+ T cells are cytotoxic lymphocytes that destroy pathogen infected and malignant cells through release of cytolytic molecules and proinflammatory cytokines. Although the role of CD8+ T cells in connective tissue diseases (CTDs) has not been explored as thoroughly as that of other immune cells, research focusing on this key component of the immune system has recently gained momentum. Aberrations in cytotoxic cell function may have implications in triggering autoimmunity and may promote tissue damage leading to exacerbation of disease. In this comprehensive review of current literature, we examine the role of CD8+ T cells in systemic lupus erythematosus, Sjögren's syndrome, systemic sclerosis, polymyositis, and dermatomyositis with specific focus on comparing what is known about CD8+ T cell peripheral blood phenotypes, CD8+ T cell function, and CD8+ T cell organ-specific profiles in adult and juvenile forms of these disorders. Although, the precise role of CD8+ T cells in the initiation of autoimmunity and disease progression remains to be elucidated, increasing evidence indicates that CD8+ T cells are emerging as an attractive target for therapy in CTDs.
Collapse
Affiliation(s)
- Anna Radziszewska
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Zachary Moulder
- University College London Medical School, University College London, London WC1E 6DE, UK
| | - Elizabeth C. Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London (UCL), University College London Hospital (UCLH), Great Ormond Street Hospital (GOSH), London WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| |
Collapse
|
31
|
Pérez-Jeldres T, Pizarro B, Ascui G, Orellana M, Cerda-Villablanca M, Alvares D, de la Vega A, Cannistra M, Cornejo B, Baéz P, Silva V, Arriagada E, Rivera-Nieves J, Estela R, Hernández-Rocha C, Álvarez-Lobos M, Tobar F. Ethnicity influences phenotype and clinical outcomes: Comparing a South American with a North American inflammatory bowel disease cohort. Medicine (Baltimore) 2022; 101:e30216. [PMID: 36086782 PMCID: PMC10980497 DOI: 10.1097/md.0000000000030216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/12/2022] [Indexed: 11/27/2022] Open
Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn disease (CD), has emerged as a global disease with an increasing incidence in developing and newly industrialized regions such as South America. This global rise offers the opportunity to explore the differences and similarities in disease presentation and outcomes across different genetic backgrounds and geographic locations. Our study includes 265 IBD patients. We performed an exploratory analysis of the databases of Chilean and North American IBD patients to compare the clinical phenotypes between the cohorts. We employed an unsupervised machine-learning approach using principal component analysis, uniform manifold approximation, and projection, among others, for each disease. Finally, we predicted the cohort (North American vs Chilean) using a random forest. Several unsupervised machine learning methods have separated the 2 main groups, supporting the differences between North American and Chilean patients with each disease. The variables that explained the loadings of the clinical metadata on the principal components were related to the therapies and disease extension/location at diagnosis. Our random forest models were trained for cohort classification based on clinical characteristics, obtaining high accuracy (0.86 = UC; 0.79 = CD). Similarly, variables related to therapy and disease extension/location had a high Gini index. Similarly, univariate analysis showed a later CD age at diagnosis in Chilean IBD patients (37 vs 24; P = .005). Our study suggests a clinical difference between North American and Chilean IBD patients: later CD age at diagnosis with a predominantly less aggressive phenotype (39% vs 54% B1) and more limited disease, despite fewer biological therapies being used in Chile for both diseases.
Collapse
Affiliation(s)
- Tamara Pérez-Jeldres
- Department of Gastroenterology, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago, Chile
- Instituto Chileno-Japonés, University of Chile, Santiago, Chile
| | - Benjamín Pizarro
- Radiology Department, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Gabriel Ascui
- La Jolla Institute for Allergy and Immunology, San Diego, CA
| | - Matías Orellana
- Department of Computer Science, Faculty of Physical Sciences and Mathematics of the University of Chile, Santiago, Chile
| | - Mauricio Cerda-Villablanca
- Integrative Biology Program, Institute of Biomedical Sciences, Center for Medical Informatics and Telemedicine, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Danilo Alvares
- Department of Statistics, Pontifical Catholic University of Chile, Santiago, Chile
| | | | - Macarena Cannistra
- Department of Gastroenterology, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago, Chile
| | - Bárbara Cornejo
- Department of Gastroenterology, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago, Chile
| | - Pablo Baéz
- Integrative Biology Program, Institute of Biomedical Sciences, Center for Medical Informatics and Telemedicine, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Verónica Silva
- Instituto Chileno-Japonés, University of Chile, Santiago, Chile
| | | | - Jesús Rivera-Nieves
- Inflammatory Bowel Disease Center, Division of Gastroenterology, University of California, San Diego, La Jolla, CA
| | - Ricardo Estela
- Instituto Chileno-Japonés, University of Chile, Santiago, Chile
| | - Cristián Hernández-Rocha
- Department of Gastroenterology, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago, Chile
| | - Manuel Álvarez-Lobos
- Department of Gastroenterology, Faculty of Medicine, Pontifical Catholic University of Chile, Santiago, Chile
| | - Felipe Tobar
- Initiative for Data & Artificial Intelligence, University of Chile
- Center for Mathematical Modeling, University of Chile, Santiago, Chile
| |
Collapse
|
32
|
Recent advances in the use of machine learning and artificial intelligence to improve diagnosis, predict flares, and enrich clinical trials in lupus. Curr Opin Rheumatol 2022; 34:374-381. [PMID: 36001343 DOI: 10.1097/bor.0000000000000902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Machine learning is a computational tool that is increasingly used for the analysis of medical data and has provided the promise of more personalized care. RECENT FINDINGS The frequency with which machine learning analytics are reported in lupus research is comparable with that of rheumatoid arthritis and cancer, yet the clinical application of these computational tools has yet to be translated into better care. Considerable work has been applied to the development of machine learning models for lupus diagnosis, flare prediction, and classification of disease using histology or other medical images, yet few models have been tested in external datasets and independent centers. Application of machine learning has yet to be reported for lupus clinical trial enrichment and automated identification of eligible patients. Integration of machine learning into lupus clinical care and clinical trials would benefit from collaborative development between clinicians and data scientists. SUMMARY Although the application of machine learning to lupus data is at a nascent stage, initial results suggest a promising future.
Collapse
|
33
|
Feng Y, Li Z, Xie C, Lu F. Correlation between peripheral blood lymphocyte subpopulations and primary systemic lupus erythematosus. Open Life Sci 2022; 17:839-845. [PMID: 36045722 PMCID: PMC9372708 DOI: 10.1515/biol-2022-0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 11/15/2022] Open
Abstract
This study explored the correlation between peripheral blood CD3+, CD3+/CD4+, CD3+/CD8+, CD4+/CD8+, CD3-/CD16+ CD56+, and CD3-CD19+ and disease activity of different subtypes of systemic lupus erythematosus (SLE). The percentages of CD3+, CD3+/CD4+, CD3+/CD8+, CD4+/CD8+, CD3-/CD16+ CD56+, and CD3-CD19+ in the peripheral blood of patients (n = 80) classified into lupus nephritis, blood involvement, and joint involvement and SLE in different active stages were detected by flow cytometry. Their correlations with baseline clinical experimental indicators of SLE patients' SLE disease activity index score (SLEDAI) and complement C3 were analyzed. The results showed that CD3+, CD3+/CD4+, and CD3+/CD8+ at baseline level were negatively correlated with SLEDAI scores. These were positively correlated with C3. In conclusion, T-lymphocyte subpopulations are closely related to SLE activity and can be used as reference indicators to evaluate the SLE activity.
Collapse
Affiliation(s)
- Yan Feng
- Rheumatology Department, First Affiliated Hospital of Anhui University of Science and Technology, Huai Nan, China
| | - Zhijun Li
- Rheumatology Department, First Affiliated Hospital of Bengbu Medical College, Bangbu, China
| | - Changhao Xie
- Rheumatology Department, First Affiliated Hospital of Bengbu Medical College, Bangbu, China
| | - Fanglin Lu
- Rheumatology Department, First Affiliated Hospital of Anhui University of Science and Technology, Huai Nan, China
| |
Collapse
|
34
|
Wang H, Lan L, Chen J, Xiao L, Han F. Peripheral blood T-cell subset and its clinical significance in lupus nephritis patients. Lupus Sci Med 2022; 9:9/1/e000717. [PMID: 35973743 PMCID: PMC9386235 DOI: 10.1136/lupus-2022-000717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 08/05/2022] [Indexed: 11/29/2022]
Abstract
Objectives Lupus nephritis (LN) is a common and severe manifestation of SLE. Memory T (TM) cells have been implicated in the pathogenesis of SLE. This study aimed to investigate the clinical significance of T-cell subsets in a cohort of patients with LN. Method The peripheral blood T cells of 24 patients with LN and 13 patients with idiopathic membranous nephropathy (iMN) were analysed by flow cytometry. SLE disease activity was evaluated by SLE Disease Activity Index-2000 (SLEDAI-2K). Patients with LN were followed up for >6 months. Results Patients with LN presented lower frequency of CD4+ cells and higher percentage of CD8+ cells than patients with iMN, which was primarily due to lower CD4+ cell count. Interestingly, patients with LN under immunosuppressants had lower CD8+CD45RO+ TM frequency (p=0.007), fewer regulatory CD4+ T cells (p=0.04) than those without immunosuppressants. Most CD4+ and CD8+ TM cells in patients with LN showed an effector memory (CD45RO+CCR7+) phenotype. The frequency of CD8+CD45RO+ TM cells among the CD8+ T cells was negatively correlated with white blood cell count, haemoglobin, platelet and serum levels of complements C3 and C4, but was positively correlated with serum IgG, erythrocyte sedimentation rate and SLEDAI score (p<0.05 each). Consistently, the frequency of CD8+CD45RO+ TM cells was higher in patients with LN with positive antidouble-stranded DNA antibody, active renal disease, extrarenal manifestations and with sclerotic glomeruli or moderate-to-severe mesangial hypercellularity in renal pathology (p<0.05). Additionally, CD8+CD45RO+ TM cell frequency was significantly lower in patients with LN with renal complete remission than that in non-remission LN (18.7% vs 31.2%, p<0.05). None of these significant correlations was observed in CD4+ TM cells. Conclusion The frequency of CD8+ TM cells correlates with disease activity and treatment response to immunosuppressant in patients with LN. CD8+ TM monitoring in patients with LN could provide more helpful indices for the monitoring and management of this disease.
Collapse
Affiliation(s)
- Huijing Wang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, China
| | - Lan Lan
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, China
| | - Liang Xiao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China .,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, China
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China .,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, China.,Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, China
| |
Collapse
|
35
|
Yamada J, Peracchi OA, Terreri MT, de Moraes-Pinto MI. Cell activation, PD-1 expression and in vitro cytokine production in patients with juvenile systemic lupus erythematosus. Lupus 2022; 31:1237-1244. [PMID: 35849633 DOI: 10.1177/09612033221112809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Juvenile systemic lupus erythematosus (jSLE) is known to be more severe and with a higher frequency of renal and central nervous system impairment when compared to systemic lupus erythematosus in adults. The study of immunological characteristics of jSLE patients might help to envisage better treatment strategies to reduce the burden of the disease. OBJECTIVE To characterize peripheral lymphocytes, assessing activation markers, and PD-1 expression on T cells; to evaluate in vitro cytokine expression upon stimulation in jSLE patients and age-matched controls. METHODOLOGY Eighteen jSLE patients on low disease activity and 25 matched healthy adolescents were evaluated for immune activation and PD-1 expression on peripheral blood lymphocytes by flow cytometry. Twenty-one cytokines were assessed by X-MAP technology after in vitro stimulation of peripheral blood with phytohemagglutinin. RESULTS jSLE patients had lower numbers of CD4 T, CD8 T, B, and NK cells; higher central memory CD8 T cell percentages were noted in jSLE adolescents in comparison with controls (p = 0.014). B cells subsets showed a higher percentage of exhausted memory subset than controls (p = 0.014). The expression of PD-1 on CD4 T and CD8 T cells did not show relevant changes in jSLE adolescents. After stimulation of peripheral blood, cell supernatant of jSLE patients showed a trend to lower concentrations of IL-10 (p=0.080) and higher concentrations of IL-23 (p = 0.063) than controls. CONCLUSIONS jSLE patients on low disease activity maintain lymphopenia of all subsets, with a B cell profile of exhaustion. Upon in vitro stimulation, peripheral blood cell supernatant showed a shift to IL-23, suggesting a role of inhibitors of this cytokine as another potential therapeutic target for those patients.
Collapse
Affiliation(s)
- Juliana Yamada
- Research Laboratory, Division of Pediatric Infectious Diseases, Department of Pediatrics, 28105Universidade Federal de São Paulo, São Paulo, Brazil
| | - Octávio Ab Peracchi
- Unit of Pediatric Rheumatology, Department of Pediatrics, 28105Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria T Terreri
- Unit of Pediatric Rheumatology, Department of Pediatrics, 28105Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria Isabel de Moraes-Pinto
- Research Laboratory, Division of Pediatric Infectious Diseases, Department of Pediatrics, 28105Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
36
|
Guthridge JM, Wagner CA, James JA. The promise of precision medicine in rheumatology. Nat Med 2022; 28:1363-1371. [PMID: 35788174 PMCID: PMC9513842 DOI: 10.1038/s41591-022-01880-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/23/2022] [Indexed: 01/07/2023]
Abstract
Systemic autoimmune rheumatic diseases (SARDs) exhibit extensive heterogeneity in clinical presentation, disease course, and treatment response. Therefore, precision medicine - whereby treatment is tailored according to the underlying pathogenic mechanisms of an individual patient at a specific time - represents the 'holy grail' in SARD clinical care. Current strategies include treat-to-target therapies and autoantibody testing for patient stratification; however, these are far from optimal. Recent innovations in high-throughput 'omic' technologies are now enabling comprehensive profiling at multiple levels, helping to identify subgroups of patients who may taper off potentially toxic medications or better respond to current molecular targeted therapies. Such advances may help to optimize outcomes and identify new pathways for treatment, but there are many challenges along the path towards clinical translation. In this Review, we discuss recent efforts to dissect cellular and molecular heterogeneity across multiple SARDs and future directions for implementing stratification approaches for SARD treatment in the clinic.
Collapse
Affiliation(s)
- Joel M Guthridge
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Departments of Medicine and Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Catriona A Wagner
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Judith A James
- Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
- Departments of Medicine and Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
37
|
Ciurtin C, Pineda-Torra I, Jury EC, Robinson GA. CD8+ T-Cells in Juvenile-Onset SLE: From Pathogenesis to Comorbidities. Front Med (Lausanne) 2022; 9:904435. [PMID: 35801216 PMCID: PMC9254716 DOI: 10.3389/fmed.2022.904435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/26/2022] [Indexed: 11/25/2022] Open
Abstract
Diagnosis of systemic lupus erythematosus (SLE) in childhood [juvenile-onset (J) SLE], results in a more severe disease phenotype including major organ involvement, increased organ damage, cardiovascular disease risk and mortality compared to adult-onset SLE. Investigating early disease course in these younger JSLE patients could allow for timely intervention to improve long-term prognosis. However, precise mechanisms of pathogenesis are yet to be elucidated. Recently, CD8+ T-cells have emerged as a key pathogenic immune subset in JSLE, which are increased in patients compared to healthy individuals and associated with more active disease and organ involvement over time. CD8+ T-cell subsets have also been used to predict disease prognosis in adult-onset SLE, supporting the importance of studying this cell population in SLE across age. Recently, single-cell approaches have allowed for more detailed analysis of immune subsets in JSLE, where type-I IFN-signatures have been identified in CD8+ T-cells expressing high levels of granzyme K. In addition, JSLE patients with an increased cardiometabolic risk have increased CD8+ T-cells with elevated type-I IFN-signaling, activation and apoptotic pathways associated with atherosclerosis. Here we review the current evidence surrounding CD8+ T-cell dysregulation in JSLE and therapeutic strategies that could be used to reduce CD8+ T-cell inflammation to improve disease prognosis.
Collapse
Affiliation(s)
- Coziana Ciurtin
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, United Kingdom
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, Division of Medicine, University College London, London, United Kingdom
| | - Elizabeth C. Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
| | - George A. Robinson
- Centre for Rheumatology Research, Division of Medicine, University College London, London, United Kingdom
- Centre for Adolescent Rheumatology Versus Arthritis, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
38
|
Qin Y, Wang Y, Meng F, Feng M, Zhao X, Gao C, Luo J. Identification of biomarkers by machine learning classifiers to assist diagnose rheumatoid arthritis-associated interstitial lung disease. Arthritis Res Ther 2022; 24:115. [PMID: 35590341 PMCID: PMC9118651 DOI: 10.1186/s13075-022-02800-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/06/2022] [Indexed: 12/14/2022] Open
Abstract
Background This study aimed to search for blood biomarkers among the profiles of patients with RA-ILD by using machine learning classifiers and probe correlations between the markers and the characteristics of RA-ILD. Methods A total of 153 RA patients were enrolled, including 75 RA-ILD and 78 RA-non-ILD. Routine laboratory data, the levels of tumor markers and autoantibodies, and clinical manifestations were recorded. Univariate analysis, least absolute shrinkage and selection operator (LASSO), random forest (RF), and partial least square (PLS) were performed, and the receiver operating characteristic (ROC) curves were plotted. Results Univariate analysis showed that, compared to RA-non-ILD, patients with RA-ILD were older (p < 0.001), had higher white blood cell (p = 0.003) and neutrophil counts (p = 0.017), had higher erythrocyte sedimentation rate (p = 0.003) and C-reactive protein (p = 0.003), had higher levels of KL-6 (p < 0.001), D-dimer (p < 0.001), fibrinogen (p < 0.001), fibrinogen degradation products (p < 0.001), lactate dehydrogenase (p < 0.001), hydroxybutyrate dehydrogenase (p < 0.001), carbohydrate antigen (CA) 19–9 (p < 0.001), carcinoembryonic antigen (p = 0.001), and CA242 (p < 0.001), but a significantly lower albumin level (p = 0.003). The areas under the curves (AUCs) of the LASSO, RF, and PLS models attained 0.95 in terms of differentiating patients with RA-ILD from those without. When data from the univariate analysis and the top 10 indicators of the three machine learning models were combined, the most discriminatory markers were age and the KL-6, D-dimer, and CA19-9, with AUCs of 0.814 [95% confidence interval (CI) 0.731–0.880], 0.749 (95% CI 0.660–0.824), 0.749 (95% CI 0.660–0.824), and 0.727 (95% CI 0.637–0.805), respectively. When all four markers were combined, the AUC reached 0.928 (95% CI 0.865–0.968). Notably, neither the KL-6 nor the CA19-9 level correlated with disease activity in RA-ILD group. Conclusions The levels of KL-6, D-dimer, and tumor markers greatly aided RA-ILD identification. Machine learning algorithms combined with traditional biostatistical analysis can diagnose patients with RA-ILD and identify biomarkers potentially associated with the disease. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02800-2.
Collapse
Affiliation(s)
- Yan Qin
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yanlin Wang
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Fanxing Meng
- The Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Min Feng
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiangcong Zhao
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jing Luo
- Department of Rheumatology, Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
39
|
Ciurtin C, Robinson GA, Pineda-Torra I, Jury EC. Comorbidity in young patients with juvenile systemic lupus erythematosus: how can we improve management? Clin Rheumatol 2022; 41:961-964. [PMID: 35178646 DOI: 10.1007/s10067-022-06093-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/03/2022] [Accepted: 02/03/2022] [Indexed: 11/03/2022]
Affiliation(s)
- Coziana Ciurtin
- Centre for Adolescent Rheumatology Research, Division of Medicine, University College London, Rayne Building, London, WC1E 6JF, UK.
| | - George A Robinson
- Centre for Adolescent Rheumatology Research, Division of Medicine, University College London, Rayne Building, London, WC1E 6JF, UK
- Centre for Rheumatology Research, Division of Medicine, University College London, Rayne Building, London, WC1E 6JF, UK
| | - Ines Pineda-Torra
- Centre for Cardiometabolic and Vascular Science, Department of Medicine, University College London, London, WC1E 6JF, UK
| | - Elizabeth C Jury
- Centre for Rheumatology Research, Division of Medicine, University College London, Rayne Building, London, WC1E 6JF, UK
| |
Collapse
|
40
|
Lerkvaleekul B, Apiwattanakul N, Tangnararatchakit K, Jirapattananon N, Srisala S, Vilaiyuk S. Associations of lymphocyte subpopulations with clinical phenotypes and long-term outcomes in juvenile-onset systemic lupus erythematosus. PLoS One 2022; 17:e0263536. [PMID: 35130317 PMCID: PMC8820627 DOI: 10.1371/journal.pone.0263536] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Juvenile-onset systemic lupus erythematosus (JSLE) is a complex and heterogeneous immune-mediated disease. Cellular components have crucial roles in disease phenotypes and outcomes. We aimed to determine the associations of lymphocyte subsets with clinical manifestations and long-term outcomes in JSLE patients. METHODS A cohort of 60 JSLE patients provided blood samples during active disease, of whom 34 provided further samples during inactive disease. In a longitudinal study, blood samples were obtained from 49 of the JSLE patients at 0, 3, and 6 months. The healthy control (HC) group consisted of 42 age-matched children. Lymphocyte subsets were analyzed by flow cytometry. RESULTS The percentages of CD4+ T, γδ T, and NK cells were significantly decreased in JSLE patients compared with HC, while the percentages of CD8+ T, NKT, and CD19+ B cells were significantly increased. The percentage of regulatory T cells (Tregs) was significantly lower in JSLE patients with lupus nephritis (LN) than in non-LN JSLE patients and HC. The patients were stratified into high and low groups by the median frequency of each lymphocyte subset. The γδ T cells high group and NK cells high group were significantly related to mucosal ulcer. The CD4+ T cells high group was significantly associated with arthritis, and the NKT cells high group was substantially linked with autoimmune hemolytic anemia. The CD8+ T cells low group was mainly related to vasculitis, and the Tregs low group was significantly associated with LN. The percentage of Tregs was significantly increased at 6 months of follow-up, and the LN JSLE group had a lower Treg percentage than the non-LN JSLE group. Predictors of remission on therapy were high Tregs, high absolute lymphocyte count, direct Coombs test positivity, and LN absence at enrollment. CONCLUSION JSLE patients exhibited altered lymphocyte subsets, which were strongly associated with clinical phenotypes and long-term outcomes.
Collapse
Affiliation(s)
- Butsabong Lerkvaleekul
- Faculty of Medicine Ramathibodi Hospital, Division of Rheumatology, Department of Pediatrics, Mahidol University, Bangkok, Thailand
| | - Nopporn Apiwattanakul
- Faculty of Medicine Ramathibodi Hospital, Division of Infectious Disease, Department of Pediatrics, Mahidol University, Bangkok, Thailand
| | - Kanchana Tangnararatchakit
- Faculty of Medicine Ramathibodi Hospital, Division of Nephrology, Department of Pediatrics, Mahidol University, Bangkok, Thailand
| | - Nisa Jirapattananon
- Faculty of Medicine Ramathibodi Hospital, Department of Pediatrics, Mahidol University, Bangkok, Thailand
| | - Supanart Srisala
- Faculty of Medicine Ramathibodi Hospital, Research Center, Mahidol University, Bangkok, Thailand
| | - Soamarat Vilaiyuk
- Faculty of Medicine Ramathibodi Hospital, Division of Rheumatology, Department of Pediatrics, Mahidol University, Bangkok, Thailand
| |
Collapse
|
41
|
McCurdy SR, Radojcic V, Tsai HL, Vulic A, Thompson E, Ivcevic S, Kanakry CG, Powell JD, Lohman B, Adom D, Paczesny S, Cooke KR, Jones RJ, Varadhan R, Symons HJ, Luznik L. Signatures of GVHD and relapse after posttransplant cyclophosphamide revealed by immune profiling and machine learning. Blood 2022; 139:608-623. [PMID: 34657151 PMCID: PMC8796655 DOI: 10.1182/blood.2021013054] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/24/2021] [Indexed: 01/29/2023] Open
Abstract
The key immunologic signatures associated with clinical outcomes after posttransplant cyclophosphamide (PTCy)-based HLA-haploidentical (haplo) and HLA-matched bone marrow transplantation (BMT) are largely unknown. To address this gap in knowledge, we used machine learning to decipher clinically relevant signatures from immunophenotypic, proteomic, and clinical data and then examined transcriptome changes in the lymphocyte subsets that predicted major posttransplant outcomes. Kinetics of immune subset reconstitution after day 28 were similar for 70 patients undergoing haplo and 75 patients undergoing HLA-matched BMT. Machine learning based on 35 candidate factors (10 clinical, 18 cellular, and 7 proteomic) revealed that combined elevations in effector CD4+ conventional T cells (Tconv) and CXCL9 at day 28 predicted acute graft-versus-host disease (aGVHD). Furthermore, higher NK cell counts predicted improved overall survival (OS) due to a reduction in both nonrelapse mortality and relapse. Transcriptional and flow-cytometric analyses of recovering lymphocytes in patients with aGVHD identified preserved hallmarks of functional CD4+ regulatory T cells (Tregs) while highlighting a Tconv-driven inflammatory and metabolic axis distinct from that seen with conventional GVHD prophylaxis. Patients developing early relapse displayed a loss of inflammatory gene signatures in NK cells and a transcriptional exhaustion phenotype in CD8+ T cells. Using a multimodality approach, we highlight the utility of systems biology in BMT biomarker discovery and offer a novel understanding of how PTCy influences alloimmune responses. Our work charts future directions for novel therapeutic interventions after these increasingly used GVHD prophylaxis platforms. Specimens collected on NCT0079656226 and NCT0080927627 https://clinicaltrials.gov/.
Collapse
Affiliation(s)
- Shannon R McCurdy
- Abramson Cancer Center and the Division of Hematology and Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Vedran Radojcic
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Hua-Ling Tsai
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ante Vulic
- Division of Biostatistics and Bioinformatics and the Sidney Kimmel Comprehensive Cancer Center and The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elizabeth Thompson
- Division of Biostatistics and Bioinformatics and the Sidney Kimmel Comprehensive Cancer Center and The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sanja Ivcevic
- Division of Hematology and Hematologic Malignancies, Department of Internal Medicine, University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Christopher G Kanakry
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jonathan D Powell
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Brian Lohman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Djamilatou Adom
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN; and
- Department of Microbiology and Immunology and Pediatrics, Medical University of South Carolina, Charleston, SC
| | - Kenneth R Cooke
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Richard J Jones
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ravi Varadhan
- Division of Biostatistics and Bioinformatics and the Sidney Kimmel Comprehensive Cancer Center and The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Heather J Symons
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Leo Luznik
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
42
|
AIM in Rheumatology. Artif Intell Med 2022. [DOI: 10.1007/978-3-030-64573-1_179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
43
|
Pandya S, Thakur A, Saxena S, Jassal N, Patel C, Modi K, Shah P, Joshi R, Gonge S, Kadam K, Kadam P. A Study of the Recent Trends of Immunology: Key Challenges, Domains, Applications, Datasets, and Future Directions. SENSORS (BASEL, SWITZERLAND) 2021; 21:7786. [PMID: 34883787 PMCID: PMC8659723 DOI: 10.3390/s21237786] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 12/13/2022]
Abstract
The human immune system is very complex. Understanding it traditionally required specialized knowledge and expertise along with years of study. However, in recent times, the introduction of technologies such as AIoMT (Artificial Intelligence of Medical Things), genetic intelligence algorithms, smart immunological methodologies, etc., has made this process easier. These technologies can observe relations and patterns that humans do and recognize patterns that are unobservable by humans. Furthermore, these technologies have also enabled us to understand better the different types of cells in the immune system, their structures, their importance, and their impact on our immunity, particularly in the case of debilitating diseases such as cancer. The undertaken study explores the AI methodologies currently in the field of immunology. The initial part of this study explains the integration of AI in healthcare and how it has changed the face of the medical industry. It also details the current applications of AI in the different healthcare domains and the key challenges faced when trying to integrate AI with healthcare, along with the recent developments and contributions in this field by other researchers. The core part of this study is focused on exploring the most common classifications of health diseases, immunology, and its key subdomains. The later part of the study presents a statistical analysis of the contributions in AI in the different domains of immunology and an in-depth review of the machine learning and deep learning methodologies and algorithms that can and have been applied in the field of immunology. We have also analyzed a list of machine learning and deep learning datasets about the different subdomains of immunology. Finally, in the end, the presented study discusses the future research directions in the field of AI in immunology and provides some possible solutions for the same.
Collapse
Affiliation(s)
- Sharnil Pandya
- Symbiosis Institute of Technology, Symbiosis International (Deemed) University, Pune 412115, India; (A.T.); (S.S.); (N.J.); (R.J.); (S.G.); (K.K.); (P.K.)
| | - Aanchal Thakur
- Symbiosis Institute of Technology, Symbiosis International (Deemed) University, Pune 412115, India; (A.T.); (S.S.); (N.J.); (R.J.); (S.G.); (K.K.); (P.K.)
| | - Santosh Saxena
- Symbiosis Institute of Technology, Symbiosis International (Deemed) University, Pune 412115, India; (A.T.); (S.S.); (N.J.); (R.J.); (S.G.); (K.K.); (P.K.)
| | - Nandita Jassal
- Symbiosis Institute of Technology, Symbiosis International (Deemed) University, Pune 412115, India; (A.T.); (S.S.); (N.J.); (R.J.); (S.G.); (K.K.); (P.K.)
| | - Chirag Patel
- Computer Science & Engineering, Devang Patel Institute of Advance Technology and Research, Changa 388421, India;
| | - Kirit Modi
- Sankalchand Patel College of Engineering, Sankalchand Patel University, Visnagar 384315, India;
| | - Pooja Shah
- Information Technology Department, Gandhinagar Institute of Technology, Ahmedabad 382010, India;
| | - Rahul Joshi
- Symbiosis Institute of Technology, Symbiosis International (Deemed) University, Pune 412115, India; (A.T.); (S.S.); (N.J.); (R.J.); (S.G.); (K.K.); (P.K.)
| | - Sudhanshu Gonge
- Symbiosis Institute of Technology, Symbiosis International (Deemed) University, Pune 412115, India; (A.T.); (S.S.); (N.J.); (R.J.); (S.G.); (K.K.); (P.K.)
| | - Kalyani Kadam
- Symbiosis Institute of Technology, Symbiosis International (Deemed) University, Pune 412115, India; (A.T.); (S.S.); (N.J.); (R.J.); (S.G.); (K.K.); (P.K.)
| | - Prachi Kadam
- Symbiosis Institute of Technology, Symbiosis International (Deemed) University, Pune 412115, India; (A.T.); (S.S.); (N.J.); (R.J.); (S.G.); (K.K.); (P.K.)
| |
Collapse
|
44
|
Kingsmore KM, Puglisi CE, Grammer AC, Lipsky PE. An introduction to machine learning and analysis of its use in rheumatic diseases. Nat Rev Rheumatol 2021; 17:710-730. [PMID: 34728818 DOI: 10.1038/s41584-021-00708-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 02/07/2023]
Abstract
Machine learning (ML) is a computerized analytical technique that is being increasingly employed in biomedicine. ML often provides an advantage over explicitly programmed strategies in the analysis of multidimensional information by recognizing relationships in the data that were not previously appreciated. As such, the use of ML in rheumatology is increasing, and numerous studies have employed ML to classify patients with rheumatic autoimmune inflammatory diseases (RAIDs) from medical records and imaging, biometric or gene expression data. However, these studies are limited by sample size, the accuracy of sample labelling, and absence of datasets for external validation. In addition, there is potential for ML models to overfit or underfit the data and, thereby, these models might produce results that cannot be replicated in an unrelated dataset. In this Review, we introduce the basic principles of ML and discuss its current strengths and weaknesses in the classification of patients with RAIDs. Moreover, we highlight the successful analysis of the same type of input data (for example, medical records) with different algorithms, illustrating the potential plasticity of this analytical approach. Altogether, a better understanding of ML and the future application of advanced analytical techniques based on this approach, coupled with the increasing availability of biomedical data, may facilitate the development of meaningful precision medicine for patients with RAIDs.
Collapse
Affiliation(s)
| | | | - Amrie C Grammer
- AMPEL BioSolutions and RILITE Research Institute, Charlottesville, VA, USA
| | - Peter E Lipsky
- AMPEL BioSolutions and RILITE Research Institute, Charlottesville, VA, USA
| |
Collapse
|
45
|
Peng J, Jury EC, Dönnes P, Ciurtin C. Machine Learning Techniques for Personalised Medicine Approaches in Immune-Mediated Chronic Inflammatory Diseases: Applications and Challenges. Front Pharmacol 2021; 12:720694. [PMID: 34658859 PMCID: PMC8514674 DOI: 10.3389/fphar.2021.720694] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
In the past decade, the emergence of machine learning (ML) applications has led to significant advances towards implementation of personalised medicine approaches for improved health care, due to the exceptional performance of ML models when utilising complex big data. The immune-mediated chronic inflammatory diseases are a group of complex disorders associated with dysregulated immune responses resulting in inflammation affecting various organs and systems. The heterogeneous nature of these diseases poses great challenges for tailored disease management and addressing unmet patient needs. Applying novel ML techniques to the clinical study of chronic inflammatory diseases shows promising results and great potential for precision medicine applications in clinical research and practice. In this review, we highlight the clinical applications of various ML techniques for prediction, diagnosis and prognosis of autoimmune rheumatic diseases, inflammatory bowel disease, autoimmune chronic kidney disease, and multiple sclerosis, as well as ML applications for patient stratification and treatment selection. We highlight the use of ML in drug development, including target identification, validation and drug repurposing, as well as challenges related to data interpretation and validation, and ethical concerns related to the use of artificial intelligence in clinical research.
Collapse
Affiliation(s)
- Junjie Peng
- Department of Medicine, Centre for Adolescent Rheumatology Versus Arthritis, University College London, London, United Kingdom
| | - Elizabeth C. Jury
- Department of Medicine, Centre for Adolescent Rheumatology Versus Arthritis, University College London, London, United Kingdom
- Department of Medicine, Centre for Rheumatology Research, University College London, London, United Kingdom
| | | | - Coziana Ciurtin
- Department of Medicine, Centre for Adolescent Rheumatology Versus Arthritis, University College London, London, United Kingdom
| |
Collapse
|
46
|
Garcia-Carretero R, Olid-Velilla M, Perez-Torrella D, Torres-Pacho N, Darnaude-Ortiz MT, Bustamate-Zuloeta AD, Tenorio JA. Predictive modeling of hypophosphatasia based on a case series of adult patients with persistent hypophosphatasemia. Osteoporos Int 2021; 32:1815-1824. [PMID: 33619648 DOI: 10.1007/s00198-021-05885-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/09/2021] [Indexed: 10/22/2022]
Abstract
UNLABELLED Approximately half of individuals with hypophosphatasemia (low levels of serum alkaline phosphatase) have hypophosphatasia, a rare genetic disease in which patients may have stress fractures, bone and joint pain, or premature tooth loss. We developed a predictive model based on specific biomarkers of this disease to better diagnose this condition. INTRODUCTION Hypophosphatasemia is a condition in which low levels of alkaline phosphatase (ALP) are detected in the serum. Some individuals presenting with this condition may have a rare genetic disease called hypophosphatasia (HPP), which involves mineralization of the bone and teeth. Lack of awareness of HPP and its nonspecific symptoms make this genetic disease difficult to diagnose. We developed a predictive model based on biomarkers of HPP such as ALP and pyridoxal 5'-phosphate (PLP), because clinical manifestations sometimes are not recognized as symptoms of HPP. METHODS We assessed 325,000 ALP results between 2010 and 2015 to identify individuals suspected of having HPP. We performed univariate and multivariate analyses to characterize the relationship between hypophosphatasemia and HPP. Using several machine learning algorithms, we developed several models based on biomarkers and compared their performance to determine the best model. RESULTS The final cohort included 45 patients who underwent a genetic test. Half (23 patients) showed a mutation of the ALPL gene that encodes the tissue-nonspecific ALP enzyme. ALP (odds ratio [OR] 0.61, 95% confidence interval [CI] 0.3-0.8, p = 0.01) and PLP (OR 1.06, 95% CI 1.01-1.15, p = 0.04) were the only variables significantly associated with the presence of HPP. Support vector machines and logistic regression were the machine learning algorithms that provided the best predictive models in terms of classification (area under the curve 0.936 and 0.844, respectively). CONCLUSIONS Given the high probability of a misdiagnosis, its nonspecific symptoms, and a lack of awareness of serum ALP levels, it is difficult to make a clinical diagnosis of HPP. Predictive models based on biomarkers are necessary to achieve a proper diagnosis. Our proposed machine learning approaches achieved reasonable performance compared to traditional statistical methods used in biomedicine, increasing the likelihood of properly diagnosing such a rare disease as HPP.
Collapse
Affiliation(s)
- R Garcia-Carretero
- Department of Internal Medicine, Mostoles University Hospital, Rey Juan Carlos University, Madrid, Spain.
| | - M Olid-Velilla
- Department of Internal Medicine, Mostoles University Hospital, Madrid, Spain
| | - D Perez-Torrella
- Department of Laboratory of Clinical Analysis, Mostoles University Hospital, Madrid, Spain
| | - N Torres-Pacho
- Department of Internal Medicine, Mostoles University Hospital, Madrid, Spain
| | - M-T Darnaude-Ortiz
- Department of Clinical Genetics, Mostoles University Hospital, Madrid, Spain
| | | | - J-A Tenorio
- Institute of Medical and Molecular Genetics (INGEMM), La Paz University Hospital, Madrid, Spain
| |
Collapse
|
47
|
Singampalli KL, Jui E, Shani K, Ning Y, Connell JP, Birla RK, Bollyky PL, Caldarone CA, Keswani SG, Grande-Allen KJ. Congenital Heart Disease: An Immunological Perspective. Front Cardiovasc Med 2021; 8:701375. [PMID: 34434978 PMCID: PMC8380780 DOI: 10.3389/fcvm.2021.701375] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/13/2021] [Indexed: 12/28/2022] Open
Abstract
Congenital heart disease (CHD) poses a significant global health and economic burden-despite advances in treating CHD reducing the mortality risk, globally CHD accounts for approximately 300,000 deaths yearly. Children with CHD experience both acute and chronic cardiac complications, and though treatment options have improved, some remain extremely invasive. A challenge in addressing these morbidity and mortality risks is that little is known regarding the cause of many CHDs and current evidence suggests a multifactorial etiology. Some studies implicate an immune contribution to CHD development; however, the role of the immune system is not well-understood. Defining the role of the immune and inflammatory responses in CHD therefore holds promise in elucidating mechanisms underlying these disorders and improving upon current diagnostic and treatment options. In this review, we address the current knowledge coinciding CHDs with immune and inflammatory associations, emphasizing conditions where this understanding would provide clinical benefit, and challenges in studying these mechanisms.
Collapse
Affiliation(s)
- Kavya L. Singampalli
- Department of Bioengineering, Rice University, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Elysa Jui
- Department of Bioengineering, Rice University, Houston, TX, United States
| | - Kevin Shani
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, United States
| | - Yao Ning
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | | | - Ravi K. Birla
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
- Division of Congenital Heart Surgery, Departments of Surgery and Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Paul L. Bollyky
- Division of Infectious Diseases, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Christopher A. Caldarone
- Division of Congenital Heart Surgery, Departments of Surgery and Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | - Sundeep G. Keswani
- Laboratory for Regenerative Tissue Repair, Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, United States
| | | |
Collapse
|
48
|
Martin-Gutierrez L, Peng J, Thompson NL, Robinson GA, Naja M, Peckham H, Wu W, J'bari H, Ahwireng N, Waddington KE, Bradford CM, Varnier G, Gandhi A, Radmore R, Gupta V, Isenberg DA, Jury EC, Ciurtin C. Stratification of Patients With Sjögren's Syndrome and Patients With Systemic Lupus Erythematosus According to Two Shared Immune Cell Signatures, With Potential Therapeutic Implications. Arthritis Rheumatol 2021; 73:1626-1637. [PMID: 33645922 DOI: 10.1002/art.41708] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/18/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Similarities in the clinical and laboratory features of primary Sjögren's syndrome (SS) and systemic lupus erythematosus (SLE) have led to attempts to treat patients with primary SS or SLE with similar biologic therapeutics. However, the results of many clinical trials are disappointing, and no biologic treatments are licensed for use in primary SS, while only a few biologic agents are available to treat SLE patients whose disease has remained refractory to other treatments. With the aim of improving treatment selections, this study was undertaken to identify distinct immunologic signatures in patients with primary SS and patients with SLE, using a stratification approach based on immune cell endotypes. METHODS Immunophentyping of 29 immune cell subsets was performed using flow cytometry in peripheral blood from patients with primary SS (n = 45), patients with SLE (n = 29), and patients with secondary SS associated with SLE (SLE/SS) (n = 14), all of whom were considered to have low disease activity or be in clinical remission, and sex-matched healthy controls (n = 31). Data were analyzed using supervised machine learning (balanced random forest, sparse partial least squares discriminant analysis), logistic regression, and multiple t-tests. Patients were stratified by K-means clustering and clinical trajectory analysis. RESULTS Patients with primary SS and patients with SLE had a similar immunologic architecture despite having different clinical presentations and prognoses. Stratification of the combined primary SS, SLE, and SLE/SS patient cohorts by K-means cluster analysis revealed 2 endotypes, characterized by distinct immune cell profiles spanning the diagnoses. A signature of 8 T cell subsets that distinctly differentiated the 2 endotypes with high accuracy (area under the curve 0.9979) was identified in logistic regression and machine learning models. In clinical trajectory analyses, the change in damage scores and disease activity levels from baseline to 5 years differed between the 2 endotypes. CONCLUSION These findings identify an immune cell toolkit that may be useful for differentiating, with high accuracy, the immunologic profiles of patients with primary SS and patients with SLE as a way to achieve targeted therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | | | - Meena Naja
- University College London and University College London Hospitals, London, UK
| | | | | | | | | | | | | | | | | | | | - Vivek Gupta
- University College London Hospitals, London, UK
| | - David A Isenberg
- University College London and University College London Hospitals, London, UK
| | - Elizabeth C Jury
- University College London and University College London Hospitals, London, UK
| | - Coziana Ciurtin
- University College London and University College London Hospitals, London, UK
| |
Collapse
|
49
|
Więsik-Szewczyk E, Rutkowska E, Kwiecień I, Korzeniowska M, Sołdacki D, Jahnz-Różyk K. Patients with Common Variable Immunodeficiency Complicated by Autoimmune Phenomena Have Lymphopenia and Reduced Treg, Th17, and NK Cells. J Clin Med 2021; 10:3356. [PMID: 34362140 PMCID: PMC8348468 DOI: 10.3390/jcm10153356] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022] Open
Abstract
Most patients with primary immune deficiency suffer from recurrent infections; however, paradoxical autoimmune phenomena can also manifest. The aim of this study was to identify immunological markers of autoimmune phenomena associated with common variable immunodeficiency (CVID). The study included 33 adults with CVID divided into two groups: (1) those with noninfectious autoimmune complications (CVID-C (n = 24)) and (2) those with only infectious symptoms (CVID-OI (n = 9)). Flow cytometry of peripheral blood was performed and compared with systemic lupus erythematosus (SLE) patients (n = 17) and healthy controls (n = 20). We found that all lymphocytes were lower in CVID-C and SLE. NK cells were lowest in CVID-C. Th17 cells were significantly reduced in CVID-C and SLE. Tregs were significantly lower in CVID-C and SLE. Bregs did not significantly differ between any groups. Class-switched memory B cells were significantly lower in CVID-C and CVID-OI. Lastly, plasmablasts were significantly higher in SLE. Among the T cell subsets, CVID-C patients had lower naive and recent thymic emigrant CD4+ T cells. In conclusion, reduced Treg, Th17, and NK cells are features of CVID with autoimmune complications, and class-switched memory B cells can help distinguish patients with different causes of autoimmunity. Future studies are needed to confirm whether reductions of Treg, Th17, and NK cells might be a biomarker of more complicated CVID cases.
Collapse
Affiliation(s)
- Ewa Więsik-Szewczyk
- Department of Internal Medicine, Pulmonology, Allergy and Clinical Immunology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (M.K.); (D.S.); (K.J.-R.)
| | - Elżbieta Rutkowska
- Laboratory of Hematology and Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (E.R.); (I.K.)
| | - Iwona Kwiecień
- Laboratory of Hematology and Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (E.R.); (I.K.)
| | - Marcelina Korzeniowska
- Department of Internal Medicine, Pulmonology, Allergy and Clinical Immunology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (M.K.); (D.S.); (K.J.-R.)
| | - Dariusz Sołdacki
- Department of Internal Medicine, Pulmonology, Allergy and Clinical Immunology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (M.K.); (D.S.); (K.J.-R.)
- Department of Clinical Immunology, Medical University of Warsaw, 02-691 Warsaw, Poland
| | - Karina Jahnz-Różyk
- Department of Internal Medicine, Pulmonology, Allergy and Clinical Immunology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (M.K.); (D.S.); (K.J.-R.)
| |
Collapse
|
50
|
Greenan-Barrett J, Doolan G, Shah D, Virdee S, Robinson GA, Choida V, Gak N, de Gruijter N, Rosser E, Al-Obaidi M, Leandro M, Zandi MS, Pepper RJ, Salama A, Jury EC, Ciurtin C. Biomarkers Associated with Organ-Specific Involvement in Juvenile Systemic Lupus Erythematosus. Int J Mol Sci 2021; 22:7619. [PMID: 34299237 PMCID: PMC8306911 DOI: 10.3390/ijms22147619] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/16/2022] Open
Abstract
Juvenile systemic lupus erythematosus (JSLE) is characterised by onset before 18 years of age and more severe disease phenotype, increased morbidity and mortality compared to adult-onset SLE. Management strategies in JSLE rely heavily on evidence derived from adult-onset SLE studies; therefore, identifying biomarkers associated with the disease pathogenesis and reflecting particularities of JSLE clinical phenotype holds promise for better patient management and improved outcomes. This narrative review summarises the evidence related to various traditional and novel biomarkers that have shown a promising role in identifying and predicting specific organ involvement in JSLE and appraises the evidence regarding their clinical utility, focusing in particular on renal biomarkers, while also emphasising the research into cardiovascular, haematological, neurological, skin and joint disease-related JSLE biomarkers, as well as genetic biomarkers with potential clinical applications.
Collapse
Affiliation(s)
- James Greenan-Barrett
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Georgia Doolan
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Devina Shah
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Simrun Virdee
- Department of Ophthalmology, Royal Free Hospital, London NW3 2QG, UK;
| | - George A. Robinson
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Varvara Choida
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Nataliya Gak
- Department of Rheumatology, University College London Hospital NHS Foundation Trust, London NW1 2BU, UK; (N.G.); (M.L.)
| | - Nina de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Elizabeth Rosser
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
| | - Muthana Al-Obaidi
- Department of Paediatric Rheumatology, Great Ormond Street Hospital, London WC1N 3JH, UK;
- NIHR Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Maria Leandro
- Department of Rheumatology, University College London Hospital NHS Foundation Trust, London NW1 2BU, UK; (N.G.); (M.L.)
- Centre for Rheumatology, Division of Medicine, University College London, London WC1E 6DH, UK;
| | - Michael S. Zandi
- Department of Neurology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London NW1 2BU, UK;
| | - Ruth J. Pepper
- Department of Renal Medicine, Royal Free Hospital, University College London, London NW3 2QG, UK; (R.J.P.); (A.S.)
| | - Alan Salama
- Department of Renal Medicine, Royal Free Hospital, University College London, London NW3 2QG, UK; (R.J.P.); (A.S.)
| | - Elizabeth C. Jury
- Centre for Rheumatology, Division of Medicine, University College London, London WC1E 6DH, UK;
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, London WC1E 6DH, UK; (J.G.-B.); (G.D.); (D.S.); (G.A.R.); (V.C.); (N.d.G.); (E.R.)
- Department of Rheumatology, University College London Hospital NHS Foundation Trust, London NW1 2BU, UK; (N.G.); (M.L.)
| |
Collapse
|