1
|
Naskar S, Sharma S, Kuotsu K, Halder S, Pal G, Saha S, Mondal S, Biswas UK, Jana M, Bhattacharjee S. The biomedical applications of artificial intelligence: an overview of decades of research. J Drug Target 2025; 33:717-748. [PMID: 39744873 DOI: 10.1080/1061186x.2024.2448711] [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: 10/31/2024] [Revised: 12/13/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
A significant area of computer science called artificial intelligence (AI) is successfully applied to the analysis of intricate biological data and the extraction of substantial associations from datasets for a variety of biomedical uses. AI has attracted significant interest in biomedical research due to its features: (i) better patient care through early diagnosis and detection; (ii) enhanced workflow; (iii) lowering medical errors; (v) lowering medical costs; (vi) reducing morbidity and mortality; (vii) enhancing performance; (viii) enhancing precision; and (ix) time efficiency. Quantitative metrics are crucial for evaluating AI implementations, providing insights, enabling informed decisions, and measuring the impact of AI-driven initiatives, thereby enhancing transparency, accountability, and overall impact. The implementation of AI in biomedical fields faces challenges such as ethical and privacy concerns, lack of awareness, technology unreliability, and professional liability. A brief discussion is given of the AI techniques, which include Virtual screening (VS), DL, ML, Hidden Markov models (HMMs), Neural networks (NNs), Generative models (GMs), Molecular dynamics (MD), and Structure-activity relationship (SAR) models. The study explores the application of AI in biomedical fields, highlighting its enhanced predictive accuracy, treatment efficacy, diagnostic efficiency, faster decision-making, personalised treatment strategies, and precise medical interventions.
Collapse
Affiliation(s)
- Sweet Naskar
- Department of Pharmaceutics, Institute of Pharmacy, Kalyani, West Bengal, India
| | - Suraj Sharma
- Department of Pharmaceutics, Sikkim Professional College of Pharmaceutical Sciences, Sikkim, India
| | - Ketousetuo Kuotsu
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, West Bengal, India
| | - Suman Halder
- Medical Department, Department of Indian Railway, Kharagpur Division, Kharagpur, West Bengal, India
| | - Goutam Pal
- Service Dispensary, ESI Hospital, Hoogly, West Bengal, India
| | - Subhankar Saha
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, West Bengal, India
| | - Shubhadeep Mondal
- Department of Pharmacology, Momtaz Begum Pharmacy College, Rajarhat, West Bengal, India
| | - Ujjwal Kumar Biswas
- School of Pharmaceutical Science (SPS), Siksha O Anusandhan (SOA) University, Bhubaneswar, Odisha, India
| | - Mayukh Jana
- School of Pharmacy, Centurion University of Technology and Management, Centurion University, Bhubaneswar, Odisha, India
| | - Sunirmal Bhattacharjee
- Department of Pharmaceutics, Bharat Pharmaceutical Technology, Amtali, Agartala, Tripura, India
| |
Collapse
|
2
|
Yu S, Wang C, Ouyang J, Luo T, Zeng F, Zhang Y, Gao L, Huang S, Wang X. Identification of candidate biomarkers correlated with the pathogenesis of breast cancer patients. Sci Rep 2025; 15:8770. [PMID: 40082607 PMCID: PMC11906855 DOI: 10.1038/s41598-025-93208-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
Breast cancer (BC) is the second leading cause of cancer-related death in females, followed by lung cancer. Disadvantages exist in conventional diagnostic techniques of BC, such as radiation risk. The present study integrated bioinformatics analysis with machine learning to elucidate potential key candidate genes associated with the tumorigenesis of BC. Eleven datasets were downloaded from the Gene Expression Omnibus (GEO) database and were consolidated into two independent cohorts (training cohort and validation cohort) after batch-effect removal. We employed "limma" package to screen differentially expressed genes (DEGs) between BC and adjacent normal breast samples. Subsequently, the most reliable diagnostic indicators were identified utilizing LASSO-Logistic regression, SVM-RFE and multivariate stepwise Logistic regression analysis. Logistic model and nomogram were created based on these hub genes and applied in external validation cohort to verify the robustness of the model. As a result, a total of six hub genes connected with BC pathogenesis were identified, including CD300LG, IGSF10, FAM83D, MAMDC2, COMP and SEMA3G. Then, a diagnostic model of BC on the basis of these genes was established. ROC analysis of the diagnostic model illustrated that AUC of the training cohort was 0.978 (0.962, 0.995). In the validation cohort, AUC of training set and validation set were 0.936 (0.910, 0.961) and 0.921 (0.870, 0.972), respectively. This indicated that the model was reliable in separating BC patients from healthy individuals. The model may assist in early diagnosis of BC with implications for improving the prognosis of BC patients.
Collapse
Affiliation(s)
- Shiqun Yu
- Yunfu Center for Disease Control and Prevention, Yunfu, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences , Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Chengman Wang
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, 330006, China
| | - Jin Ouyang
- Jiangxi Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences , Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Ting Luo
- Infection Control Center, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences , Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Fanfan Zeng
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, 330006, China
| | - Yu Zhang
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, 330006, China
| | - Liyun Gao
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences , Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Shaoxin Huang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences , Jiujiang University, Jiujiang, 332005, Jiangxi, China
| | - Xin Wang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences , Jiujiang University, Jiujiang, 332005, Jiangxi, China.
| |
Collapse
|
3
|
Pang Y, Nguyen WQ, Guerrero LI, Chrisman LP, Hooper MJ, McCarthy MC, Hales MK, Lipman RE, Paller AS, Guitart J, Zhou XA. Deciphering the Etiologies of Adult Erythroderma: An Updated Guide to Presentations, Diagnostic Tools, Pathophysiologies, and Treatments. Am J Clin Dermatol 2024; 25:927-950. [PMID: 39348008 DOI: 10.1007/s40257-024-00886-9] [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] [Accepted: 08/21/2024] [Indexed: 10/01/2024]
Abstract
Erythroderma, an inflammatory skin condition characterized by widespread erythema with variable degrees of exfoliation, pustulation, or vesiculobullous formation, is associated with high morbidity and mortality. Determining the underlying cause of erythroderma frequently presents a diagnostic challenge, which may contribute to the condition's relatively poor prognosis. This review covers the clinical presentation, pathophysiology, diagnosis, and treatment of erythroderma. It discusses similarities and differences among the many underlying etiologies of the condition and differences between erythrodermic and non-erythrodermic presentations of the same dermatosis. Finally, this article explores current research that may provide future tools in the diagnosis and management of erythroderma.
Collapse
Affiliation(s)
- Yanzhen Pang
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA
| | - William Q Nguyen
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Liliana I Guerrero
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA
| | - Lauren P Chrisman
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA
| | - Madeline J Hooper
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA
| | - Morgan C McCarthy
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA
| | - Molly K Hales
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA
| | - Rachel E Lipman
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA
| | - Amy S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA
| | - Joan Guitart
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA
| | - Xiaolong A Zhou
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 676 N Saint Clair, Arkes 1600, Chicago, IL, 60611, USA.
| |
Collapse
|
4
|
Barlow R, Bewley A, Gkini MA. AI in Psoriatic Disease: Scoping Review. JMIR DERMATOLOGY 2024; 7:e50451. [PMID: 39413371 PMCID: PMC11525079 DOI: 10.2196/50451] [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: 06/30/2023] [Revised: 12/09/2023] [Accepted: 07/11/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Artificial intelligence (AI) has many applications in numerous medical fields, including dermatology. Although the majority of AI studies in dermatology focus on skin cancer, there is growing interest in the applicability of AI models in inflammatory diseases, such as psoriasis. Psoriatic disease is a chronic, inflammatory, immune-mediated systemic condition with multiple comorbidities and a significant impact on patients' quality of life. Advanced treatments, including biologics and small molecules, have transformed the management of psoriatic disease. Nevertheless, there are still considerable unmet needs. Globally, delays in the diagnosis of the disease and its severity are common due to poor access to health care systems. Moreover, despite the abundance of treatments, we are unable to predict which is the right medication for the right patient, especially in resource-limited settings. AI could be an additional tool to address those needs. In this way, we can improve rates of diagnosis, accurately assess severity, and predict outcomes of treatment. OBJECTIVE This study aims to provide an up-to-date literature review on the use of AI in psoriatic disease, including diagnostics and clinical management as well as addressing the limitations in applicability. METHODS We searched the databases MEDLINE, PubMed, and Embase using the keywords "AI AND psoriasis OR psoriatic arthritis OR psoriatic disease," "machine learning AND psoriasis OR psoriatic arthritis OR psoriatic disease," and "prognostic model AND psoriasis OR psoriatic arthritis OR psoriatic disease" until June 1, 2023. Reference lists of relevant papers were also cross-examined for other papers not detected in the initial search. RESULTS Our literature search yielded 38 relevant papers. AI has been identified as a key component in digital health technologies. Within this field, there is the potential to apply specific techniques such as machine learning and deep learning to address several aspects of managing psoriatic disease. This includes diagnosis, particularly useful for remote teledermatology via photographs taken by patients as well as monitoring and estimating severity. Similarly, AI can be used to synthesize the vast data sets already in place through patient registries which can help identify appropriate biologic treatments for future cohorts and those individuals most likely to develop complications. CONCLUSIONS There are multiple advantageous uses for AI and digital health technologies in psoriatic disease. With wider implementation of AI, we need to be mindful of potential limitations, such as validation and standardization or generalizability of results in specific populations, such as patients with darker skin phototypes.
Collapse
Affiliation(s)
- Richard Barlow
- Dermatology Department, University Hospital Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Anthony Bewley
- Department of Dermatology, The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Maria Angeliki Gkini
- Department of Dermatology, The Royal London Hospital, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
5
|
Ravipati A, Elman SA. The state of artificial intelligence for systemic dermatoses: Background and applications for psoriasis, systemic sclerosis, and much more. Clin Dermatol 2024; 42:487-491. [PMID: 38909858 DOI: 10.1016/j.clindermatol.2024.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Artificial intelligence (AI) has been steadily integrated into dermatology, with AI platforms already attempting to identify skin cancers and diagnose benign versus malignant lesions. Although not as widely known, AI programs have also been utilized as diagnostic and prognostic tools for dermatologic conditions with systemic or extracutaneous involvement, especially for diseases with autoimmune etiologies. We have provided a primer on commonly used AI platforms and the practical applicability of these algorithms in dealing with psoriasis, systemic sclerosis, and dermatomyositis as a microcosm for future directions in the field. With a rapidly changing landscape in dermatology and medicine as a whole, AI could be a versatile tool to support clinicians and enhance access to care.
Collapse
Affiliation(s)
- Advaitaa Ravipati
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Scott A Elman
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
6
|
Trivedi A, Bose D, Moffat K, Pearson E, Walsh D, Cohen D, Skupsky J, Chao L, Golier J, Janulewicz P, Sullivan K, Krengel M, Tuteja A, Klimas N, Chatterjee S. Gulf War Illness Is Associated with Host Gut Microbiome Dysbiosis and Is Linked to Altered Species Abundance in Veterans from the BBRAIN Cohort. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:1102. [PMID: 39200711 PMCID: PMC11354743 DOI: 10.3390/ijerph21081102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024]
Abstract
Gulf War Illness (GWI) is a debilitating condition marked by chronic fatigue, cognitive problems, pain, and gastrointestinal (GI) complaints in veterans who were deployed to the 1990-1991 Gulf War. Fatigue, GI complaints, and other chronic symptoms continue to persist more than 30 years post-deployment. Several potential mechanisms for the persistent illness have been identified and our prior pilot study linked an altered gut microbiome with the disorder. This study further validates and builds on our prior preliminary findings of host gut microbiome dysbiosis in veterans with GWI. Using stool samples and Multidimensional Fatigue Inventory (MFI) data from 89 GW veteran participants (63 GWI cases and 26 controls) from the Boston biorepository, recruitment, and integrative network (BBRAIN) for Gulf War Illness, we found that the host gut bacterial signature of veterans with GWI showed significantly different Bray-Curtis beta diversity than control veterans. Specifically, a higher Firmicutes to Bacteroidetes ratio, decrease in Akkermansia sp., Bacteroides thetaiotamicron, Bacteroides fragilis, and Lachnospiraceae genera and increase in Blautia, Streptococcus, Klebsiella, and Clostridium genera, that are associated with gut, immune, and brain health, were shown. Further, using MaAsLin and Boruta algorithms, Coprococcus and Eisenbergiella were identified as important predictors of GWI with an area under the curve ROC predictive value of 74.8%. Higher self-reported MFI scores in veterans with GWI were also significantly associated with an altered gut bacterial diversity and species abundance of Lachnospiraceae and Blautia. These results suggest potential therapeutic targets for veterans with GWI that target the gut microbiome and specific symptoms of the illness.
Collapse
Affiliation(s)
- Ayushi Trivedi
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (A.T.); (D.B.)
| | - Dipro Bose
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (A.T.); (D.B.)
| | - Kelly Moffat
- CosmosID, Germantown, MD 20874, USA; (K.M.); (D.W.)
| | | | - Dana Walsh
- CosmosID, Germantown, MD 20874, USA; (K.M.); (D.W.)
| | - Devra Cohen
- Miami VA Healthcare System, Miami, FL 33125, USA;
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
| | - Jonathan Skupsky
- VA Research and Development, VA Long Beach Health Care, Long Beach, CA 90822, USA;
| | - Linda Chao
- San Francisco Veterans Affairs Health Care System, San Francisco, CA 94121, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA 94143, USA
| | - Julia Golier
- J. Peters VA Medical Center, Bronx, NY 10468, USA;
- Psychiatry Department, Icahn School of Medicine at Mount Sinai, 1428 Madison Ave, New York, NY 10029, USA
| | - Patricia Janulewicz
- Department of Environmental Health, Boston University School of Public Health, 715 Albany St. T4W, Boston, MA 02130, USA; (P.J.)
| | - Kimberly Sullivan
- Department of Environmental Health, Boston University School of Public Health, 715 Albany St. T4W, Boston, MA 02130, USA; (P.J.)
| | - Maxine Krengel
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02130, USA;
| | - Ashok Tuteja
- Division of Gastroenterology, School of Medicine, University of Utah, Salt Lake City, UT 84132, USA;
| | - Nancy Klimas
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
- Geriatric Research and Education Clinical Center, Miami VA Heathcare System, Miami, FL 33125, USA
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental and Occupational Health, Program in Public Health, Susan and Henry Samueli College of Health Sciences, University of California, Irvine, CA 92697, USA; (A.T.); (D.B.)
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
- Department of Medicine, Infectious Disease, UCI School of Medicine, Irvine, CA 92697, USA
| |
Collapse
|
7
|
Wang X, Sun J, Zhang X, Chen W, Cao J, Hu H. Metagenomics reveals unique gut mycobiome biomarkers in psoriasis. Skin Res Technol 2024; 30:e13822. [PMID: 38970783 PMCID: PMC11227279 DOI: 10.1111/srt.13822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/04/2024] [Indexed: 07/08/2024]
Abstract
PURPOSE In present, the diagnosis of psoriasis is mainly based on the patient's typical clinical manifestations, dermoscopy and skin biopsy, and unlike other immune diseases, psoriasis lacks specific indicators in the blood. Therefore, we are required to search novel biomarkers for the diagnosis of psoriasis. METHODS In this study, we analyzed the composition and the differences of intestinal fungal communities composition between psoriasis patients and healthy individuals in order to find the intestinal fungal communities associated with the diagnosis of psoriasis. We built a machine learning model and identified potential microbial markers for the diagnosis of psoriasis. RESULTS The results of AUROC (area under ROC) showed that Aspergillus puulaauensis (AUROC = 0.779), Kazachstania africana (AUROC = 0.750) and Torulaspora delbrueckii (AUROC = 0.745) had high predictive ability (AUROC > 0.7) for predicting psoriasis, While Fusarium keratoplasticum (AUROC = 0.670) was relatively lower (AUROC < 0.7). CONCLUSION The strategy based on the prediction of intestinal fungal communities provides a new idea for the diagnosis of psoriasis and is expected to become an auxiliary diagnostic method for psoriasis.
Collapse
Affiliation(s)
- Xuan Wang
- Department of DermatologyLianyungang Oriental HospitalLianyungangChina
| | - Jiaxin Sun
- Department of DermatologyLianyungang Oriental HospitalLianyungangChina
| | - Xiandan Zhang
- Department of Gynecology and ObstetricsShenzhen Hospital of University of Hong KongShenzhenChina
| | - Wei Chen
- Department of DermatologyLianyungang Oriental HospitalLianyungangChina
| | - Jing Cao
- Department of DermatologyLianyungang Oriental HospitalLianyungangChina
| | - Huimin Hu
- Department of DermatologyThe Affiliated Huai’an Hospital of Xuzhou Medical University and The Second People’s Hospital of Huai’anHuai’anChina
| |
Collapse
|
8
|
Chen C, Cai R, Zhou J, Zhang D, Chen L. GPR15LG regulates psoriasis-like inflammation by down-regulating inflammatory factors on keratinocytes. Biosci Rep 2024; 44:BSR20231347. [PMID: 38393364 PMCID: PMC11147810 DOI: 10.1042/bsr20231347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 01/09/2024] [Accepted: 02/21/2024] [Indexed: 02/25/2024] Open
Abstract
Psoriasis is a common chronic inflammatory skin disease characterized by aberrant proliferation of keratinocytes and infiltration of immune cells. We previously found that GPR15LG protein is highly expressed in psoriasis lesional skin and it positively regulates psoriatic keratinocyte proliferation. Our data also showed that GPR15LG could regulate the activity of NF-κB pathway, which is associated with psoriatic inflammation. In the present study, we demonstrated that Gpr15lg (ortholog of GPR15LG) knockdown attenuated the severity of imiquimod (IMQ)-induced psoriasis-like inflammation in mice. Such an effect was achieved by down-regulating the expression of inflammatory cytokines interleukin (IL)-1α, IL-1β, tumor necrosis factor (TNF)-α and S100A7. Consistently, GPR15LG knockdown in vitro significantly downgraded the expression of inflammatory factors in the cellular model of psoriasis. These results suggested that GPR15LG could be involved in the development of psoriasis by regulating inflammation.
Collapse
Affiliation(s)
- Caifeng Chen
- Department of Dermatology, Fujian Provincial Hospital, Clinical Medical College of Fujian Medical University, Fujian Fuzhou, China
| | - Renhui Cai
- Department of Dermatology, Fujian Provincial Hospital, Clinical Medical College of Fujian Medical University, Fujian Fuzhou, China
| | - Jun Zhou
- Department of Dermatology, Fujian Provincial Hospital, Clinical Medical College of Fujian Medical University, Fujian Fuzhou, China
| | - Danqun Zhang
- Department of Dermatology, Fujian Provincial Hospital, Clinical Medical College of Fujian Medical University, Fujian Fuzhou, China
| | - Li Chen
- Department of Dermatology, Fujian Provincial Hospital, Clinical Medical College of Fujian Medical University, Fujian Fuzhou, China
| |
Collapse
|
9
|
Wang T, Fang X, Sheng X, Li M, Mei Y, Mei Q, Pan A. Identification of immune characteristic biomarkers and therapeutic targets in cuproptosis for sepsis by integrated bioinformatics analysis and single-cell RNA sequencing analysis. Heliyon 2024; 10:e27379. [PMID: 38495196 PMCID: PMC10943398 DOI: 10.1016/j.heliyon.2024.e27379] [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: 08/12/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024] Open
Abstract
Background Cuproptosis is a copper-dependent cell death that is connected to the development and immune response of multiple diseases. However, the function of cuproptosis in the immune characteristics of sepsis remains unclear. Method We obtained two sepsis datasets (GSE9960 and GSE134347) from the GEO database and classified the raw data with R packages. Cuproptosis-related genes were manually curated, and differentially expressed cuproptosis-related genes (DECuGs) were identified. Afterwards, we applied enrichment analysis and identified key DECuGs by performing machine learning techniques. Then, the immune cell infiltrations and correlation between DECuGs and immunocyte features were created by the CIBERSORT algorithm. Subsequently, unsupervised hierarchical clustering analysis was performed based on key DECuGs. We then constructed a ceRNA network based on key DECuGs by using multi-step computational strategies and predicted potential drugs in the DrugBank database. Finally, the role of these key genes in immune cells was validated at the single-cell RNA level between septic patients and healthy controls. Results Overall, 16 DECuGs were obtained, and most of them had lower expression levels in sepsis samples. Afterwards, we obtained six key DECuGs by performing machine learning. Then, the LIPT1-T-cell CD4 memory resting was the most positively correlated DECuG-immunocyte pair. Subsequently, two different subclusters were identified by six DECuGs. Bioinformatics analysis revealed that there were different immune characteristics between the two subclusters. Moreover, we identified the key lncRNA OIP5-AS1 within the ceRNA network and obtained 4 drugs that may represent novel drugs for sepsis. Finally, these key DECuGs were statistically significantly dysregulated in another validation set and showed a major distribution in monocytes, T cells, B cells, NK cells and platelets at the single-cell RNA level. Conclusion These findings suggest that cuproptosis might promote the progression of sepsis by affecting the immune system and metabolic dysfunction, which provides a new direction for understanding potential pathogenic processes and therapeutic targets in sepsis.
Collapse
Affiliation(s)
- Tianfeng Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui Province, 230001, China
| | - Xiaowei Fang
- Department of Critical Care Medicine, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui Province, 230001, China
| | - Ximei Sheng
- WanNan Medical College, Wuhu, Anhui, 241002, China
| | - Meng Li
- Department of Intensive Care Unit, The Affiliated Provincial Hospital of Anhui Medical University, Anhui, 230001, China
| | - Yulin Mei
- WanNan Medical College, Wuhu, Anhui, 241002, China
| | - Qing Mei
- Department of Critical Care Medicine, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui Province, 230001, China
| | - Aijun Pan
- Department of Critical Care Medicine, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui Province, 230001, China
| |
Collapse
|
10
|
Liu Z, Wang X, Ma Y, Lin Y, Wang G. Artificial intelligence in psoriasis: Where we are and where we are going. Exp Dermatol 2023; 32:1884-1899. [PMID: 37740587 DOI: 10.1111/exd.14938] [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: 06/15/2023] [Revised: 09/05/2023] [Accepted: 09/09/2023] [Indexed: 09/24/2023]
Abstract
Artificial intelligence (AI) is a field of computer science that involves the development of programs designed to replicate human cognitive processes and the analysis of complex data. In dermatology, which is predominantly a visual-based diagnostic field, AI has become increasingly important in improving professional processes, particularly in the diagnosis of psoriasis. In this review, we summarized current AI applications in psoriasis: (i) diagnosis, including identification, classification, lesion segmentation, lesion severity and area scoring; (ii) treatment, including prediction treatment efficiency and prediction candidate drugs; (iii) management, including e-health and preventive medicine. Key challenges and future aspects of AI in psoriasis were also discussed, in hope of providing potential directions for future studies.
Collapse
Affiliation(s)
- Zhenhua Liu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinyu Wang
- Department of Economics, Finance and Healthcare Administration, Valdosta State University, Valdosta, Georgia, USA
| | - Yao Ma
- Student Brigade of Basic Medicine School, Fourth Military Medical University, Xi'an, China
| | - Yiting Lin
- Department of Dermatology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
11
|
Hong S, Wang H, Chan S, Zhang J, Chen B, Ma X, Chen X. Identifying Macrophage-Related Genes in Ulcerative Colitis Using Weighted Coexpression Network Analysis and Machine Learning. Mediators Inflamm 2023; 2023:4373840. [PMID: 38633005 PMCID: PMC11023725 DOI: 10.1155/2023/4373840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/23/2023] [Accepted: 09/27/2023] [Indexed: 04/19/2024] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease of unknown cause that typically affects the colon and rectum. Innate intestinal immunity, including macrophages, plays a significant role in the pathological development of UC. Using the CIBERSORT algorithm, we observed elevated levels of 22 types of immune cell infiltrates, as well as increased M1 and decreased M2 macrophages in UC compared to normal colonic mucosa. Weighted gene coexpression network analysis (WGCNA) was used to identify modules associated with macrophages and UC, resulting in the identification of 52 macrophage-related genes (MRGs) that were enriched in macrophages at single-cell resolution. Consensus clustering based on these 52 MRGs divided the integrated UC cohorts into three subtypes. Machine learning algorithms were used to identify ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), sodium- and chloride-dependent neutral and basic amino acid transporter B(0+) (SLC6A14), and 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2) in the training set, and their diagnostic value was validated in independent validation sets. Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) revealed the main biological effects, and that interleukin-17 was one of several signaling pathways enriched by the three genes. We also constructed a competitive endogenous RNA (CeRNA) network reflecting a potential posttranscriptional regulatory mechanism. Expression of diagnostic markers was validated in vivo and in biospecimens, and our immunohistochemistry (IHC) results confirmed that HMGCS2 gradually decreased during the transformation of UC to colorectal cancer. In conclusion, ENPP1, SLC6A14, and HMGCS2 are associated with macrophages and the progression of UC pathogenesis and have good diagnostic value for patients with UC.
Collapse
Affiliation(s)
- Shaocheng Hong
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
- Anhui Provincial Key Laboratory of Digestive Diseases, Hefei, China
| | - Hongqian Wang
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
- Anhui Provincial Key Laboratory of Digestive Diseases, Hefei, China
| | - Shixin Chan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Jiayi Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
- Anhui Provincial Key Laboratory of Digestive Diseases, Hefei, China
| | - Bangjie Chen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Xiaohan Ma
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
- Anhui Provincial Key Laboratory of Digestive Diseases, Hefei, China
| | - Xi Chen
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
- Anhui Provincial Key Laboratory of Digestive Diseases, Hefei, China
| |
Collapse
|
12
|
Omiye JA, Gui H, Daneshjou R, Cai ZR, Muralidharan V. Principles, applications, and future of artificial intelligence in dermatology. Front Med (Lausanne) 2023; 10:1278232. [PMID: 37901399 PMCID: PMC10602645 DOI: 10.3389/fmed.2023.1278232] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
This paper provides an overview of artificial-intelligence (AI), as applied to dermatology. We focus our discussion on methodology, AI applications for various skin diseases, limitations, and future opportunities. We review how the current image-based models are being implemented in dermatology across disease subsets, and highlight the challenges facing widespread adoption. Additionally, we discuss how the future of AI in dermatology might evolve and the emerging paradigm of large language, and multi-modal models to emphasize the importance of developing responsible, fair, and equitable models in dermatology.
Collapse
Affiliation(s)
| | - Haiwen Gui
- Department of Dermatology, Stanford University, Stanford, CA, United States
| | - Roxana Daneshjou
- Department of Dermatology, Stanford University, Stanford, CA, United States
- Department of Biomedical Data Science, Stanford University, Stanford, CA, United States
| | - Zhuo Ran Cai
- Department of Dermatology, Stanford University, Stanford, CA, United States
| | | |
Collapse
|
13
|
Kaczmarska A, Kwiatkowska D, Skrzypek KK, Kowalewski ZT, Jaworecka K, Reich A. Pathomechanism of Pruritus in Psoriasis and Atopic Dermatitis: Novel Approaches, Similarities and Differences. Int J Mol Sci 2023; 24:14734. [PMID: 37834183 PMCID: PMC10573181 DOI: 10.3390/ijms241914734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/24/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Pruritus is defined as an unpleasant sensation that elicits a desire to scratch. Nearly a third of the world's population may suffer from pruritus during their lifetime. This symptom is widely observed in numerous inflammatory skin diseases-e.g., approximately 70-90% of patients with psoriasis and almost every patient with atopic dermatitis suffer from pruritus. Although the pathogenesis of atopic dermatitis and psoriasis is different, the complex intricacies between several biochemical mediators, enzymes, and pathways seem to play a crucial role in both conditions. Despite the high prevalence of pruritus in the general population, the pathogenesis of this symptom in various conditions remains elusive. This review aims to summarize current knowledge about the pathogenesis of pruritus in psoriasis and atopic dermatitis. Each molecule involved in the pruritic pathway would merit a separate chapter or even an entire book, however, in the current review we have concentrated on some reports which we found crucial in the understanding of pruritus. However, the pathomechanism of pruritus is an extremely complex and intricate process. Moreover, many of these signaling pathways are currently undergoing detailed analysis or are still unexplained. As a result, it is currently difficult to take an objective view of how far we have come in elucidating the pathogenesis of pruritus in the described diseases. Nevertheless, considerable progress has been made in recent years.
Collapse
Affiliation(s)
- Agnieszka Kaczmarska
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-055 Rzeszów, Poland; (A.K.); (D.K.); (K.J.)
| | - Dominika Kwiatkowska
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-055 Rzeszów, Poland; (A.K.); (D.K.); (K.J.)
| | | | | | - Kamila Jaworecka
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-055 Rzeszów, Poland; (A.K.); (D.K.); (K.J.)
| | - Adam Reich
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-055 Rzeszów, Poland; (A.K.); (D.K.); (K.J.)
| |
Collapse
|
14
|
Wang T, Xu S, Zhang L, Yang T, Fan X, Zhu C, Wang Y, Tong F, Mei Q, Pan A. Identification of immune-related lncRNA in sepsis by construction of ceRNA network and integrating bioinformatic analysis. BMC Genomics 2023; 24:484. [PMID: 37620751 PMCID: PMC10464037 DOI: 10.1186/s12864-023-09535-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Sepsis is a high mortality disease which seriously threatens human life and health, for which the pathogenetic mechanism still unclear. There is increasing evidence showed that immune and inflammation responses are key players in the development of sepsis pathology. LncRNAs, which act as ceRNAs, have critical roles in various diseases. However, the regulatory roles of ceRNA in the immunopathogenesis of sepsis have not yet been elucidated. RESULTS In this study, we aimed to identify immune biomarkers associated with sepsis. We first generated a global immune-associated ceRNA (IMCE) network based on data describing interactions pairs of gene-miRNA and miRNA-lncRNA. Afterward, we excavated a dysregulated sepsis immune-associated ceRNA (SPIMC) network from the global IMCE network by means of a multi-step computational approach. Functional enrichment indicated that lncRNAs in SPIMC network have pivotal roles in the immune mechanism underlying sepsis. Subsequently, we identified module and hub genes (CD4 and STAT4) via construction of a sepsis immune-related PPI network. Then, we identified hub genes based on the modular structure of PPI network and generated a ceRNA subnetwork to analyze key lncRNAs associated with sepsis. Finally, 6 lncRNAs (LINC00265, LINC00893, NDUFA6-AS1, NOP14-AS1, PRKCQ-AS1 and ZNF674-AS1) that identified as immune biomarkers of sepsis. Moreover, the CIBERSORT algorithm and the infiltration of circulating immune cells types were performed to identify the inflammatory state of sepsis. Correlation analyses between immune cells and sepsis immune biomarkers showed that the LINC00265 was strongly positive correlated with the macrophages M2 (r = 0.77). CONCLUSION Collectively, these results may suggest that these lncRNAs (LINC00265, LINC00893, NDUFA6-AS1, NOP14-AS1, PRKCQ-AS1 and ZNF674-AS1) played important roles in the immune pathogenesis of sepsis and provide potential therapeutic targets for further researches on immune therapy treatment in patients with sepsis.
Collapse
Affiliation(s)
- Tianfeng Wang
- Department of Critical Care Medicine, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui Province, China
| | - Si Xu
- Department of Neurology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Zhang
- Department of Critical Care Medicine, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui Province, China
| | - Tianjun Yang
- Department of Critical Care Medicine, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui Province, China
| | - Xiaoqin Fan
- Department of Critical Care Medicine, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui Province, China
| | - Chunyan Zhu
- Department of Critical Care Medicine, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui Province, China
| | - Yinzhong Wang
- Department of Critical Care Medicine, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui Province, China
| | - Fei Tong
- Department of Critical Care Medicine, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui Province, China
| | - Qing Mei
- Department of Critical Care Medicine, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui Province, China.
| | - Aijun Pan
- Department of Critical Care Medicine, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui Province, China.
| |
Collapse
|
15
|
Okamoto Y, Shikano S. Emerging roles of a chemoattractant receptor GPR15 and ligands in pathophysiology. Front Immunol 2023; 14:1179456. [PMID: 37457732 PMCID: PMC10348422 DOI: 10.3389/fimmu.2023.1179456] [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: 03/04/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Chemokine receptors play a central role in the maintenance of immune homeostasis and development of inflammation by directing leukocyte migration to tissues. GPR15 is a G protein-coupled receptor (GPCR) that was initially known as a co-receptor for human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV), with structural similarity to other members of the chemoattractant receptor family. Since the discovery of its novel function as a colon-homing receptor of T cells in mice a decade ago, GPR15 has been rapidly gaining attention for its involvement in a variety of inflammatory and immune disorders. The recent identification of its natural ligand C10orf99, a chemokine-like polypeptide strongly expressed in gastrointestinal tissues, has established that GPR15-C10orf99 is a novel signaling axis that controls intestinal homeostasis and inflammation through the migration of immune cells. In addition, it has been demonstrated that C10orf99-independent functions of GPR15 and GPR15-independent activities of C10orf99 also play significant roles in the pathophysiology. Therefore, GPR15 and its ligands are potential therapeutic targets. To provide a basis for the future development of GPR15- or GPR15 ligand-targeted therapeutics, we have summarized the latest advances in the role of GPR15 and its ligands in human diseases as well as the molecular mechanisms that regulate GPR15 expression and functions.
Collapse
Affiliation(s)
| | - Sojin Shikano
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
16
|
Ye SB, Cheng YK, Li PS, Zhang L, Zhang LH, Huang Y, Chen P, Wang Y, Wang C, Peng JH, Shi LS, Ling L, Wu XJ, Qin J, Yang ZH, Lan P. High-throughput proteomics profiling-derived signature associated with chemotherapy response and survival for stage II/III colorectal cancer. NPJ Precis Oncol 2023; 7:50. [PMID: 37258779 DOI: 10.1038/s41698-023-00400-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 05/12/2023] [Indexed: 06/02/2023] Open
Abstract
Adjuvant chemotherapy (ACT) is usually used to reduce the risk of disease relapse and improve survival for stage II/III colorectal cancer (CRC). However, only a subset of patients could benefit from ACT. Thus, there is an urgent need to identify improved biomarkers to predict survival and stratify patients to refine the selection of ACT. We used high-throughput proteomics to analyze tumor and adjacent normal tissues of stage II/III CRC patients with /without relapse to identify potential markers for predicting prognosis and benefit from ACT. The machine learning approach was applied to identify relapse-specific markers. Then the artificial intelligence (AI)-assisted multiplex IHC was performed to validate the prognostic value of the relapse-specific markers and construct a proteomic-derived classifier for stage II/III CRC using 3 markers, including FHL3, GGA1, TGFBI. The proteomics profiling-derived signature for stage II/III CRC (PS) not only shows good accuracy to classify patients into high and low risk of relapse and mortality in all three cohorts, but also works independently of clinicopathologic features. ACT was associated with improved disease-free survival (DFS) and overall survival (OS) in stage II (pN0) patients with high PS and pN2 patients with high PS. This study demonstrated the clinical significance of proteomic features, which serve as a valuable source for potential biomarkers. The PS classifier provides prognostic value for identifying patients at high risk of relapse and mortality and optimizes individualized treatment strategy by detecting patients who may benefit from ACT for survival.
Collapse
Affiliation(s)
- Shu-Biao Ye
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Yi-Kan Cheng
- Department of Radiation Oncology; The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Pei-Si Li
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
- Sun Yat-sen University School of Medicine, Sun Yat-sen University, Guangzhou, PR China
| | - Lin Zhang
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Lian-Hai Zhang
- Department of Surgery, Peking University Cancer Hospital and Institute, Beijing, PR China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Ping Chen
- Department of VIP, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Yi Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing Proteome Research Center, Beijing, China
| | - Chao Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Jian-Hong Peng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Li-Shuo Shi
- Department of Probability and Statistics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Li Ling
- Department of Probability and Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, PR China
| | - Xiao-Jian Wu
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
| | - Jun Qin
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing Proteome Research Center, Beijing, China.
| | - Zi-Huan Yang
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
| | - Ping Lan
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
17
|
Lilja S, Li X, Smelik M, Lee EJ, Loscalzo J, Marthanda PB, Hu L, Magnusson M, Sysoev O, Zhang H, Zhao Y, Sjöwall C, Gawel D, Wang H, Benson M. Multi-organ single-cell analysis reveals an on/off switch system with potential for personalized treatment of immunological diseases. Cell Rep Med 2023; 4:100956. [PMID: 36858042 PMCID: PMC10040389 DOI: 10.1016/j.xcrm.2023.100956] [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: 08/17/2022] [Revised: 09/30/2022] [Accepted: 02/03/2023] [Indexed: 03/03/2023]
Abstract
Prioritization of disease mechanisms, biomarkers, and drug targets in immune-mediated inflammatory diseases (IMIDs) is complicated by altered interactions between thousands of genes. Our multi-organ single-cell RNA sequencing of a mouse IMID model, namely collagen-induced arthritis, shows highly complex and heterogeneous expression changes in all analyzed organs, even though only joints showed signs of inflammation. We organized those into a multi-organ multicellular disease model, which shows predicted molecular interactions within and between organs. That model supports that inflammation is switched on or off by altered balance between pro- and anti-inflammatory upstream regulators (URs) and downstream pathways. Meta-analyses of human IMIDs show a similar, but graded, on/off switch system. This system has the potential to prioritize, diagnose, and treat optimal combinations of URs on the levels of IMIDs, subgroups, and individual patients. That potential is supported by UR analyses in more than 600 sera from patients with systemic lupus erythematosus.
Collapse
Affiliation(s)
- Sandra Lilja
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Mavatar, Inc, Vasagatan, 11120 Stockholm, Sweden
| | - Xinxiu Li
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Martin Smelik
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Eun Jung Lee
- Department of Otorhinolaryngology, Yonsei University Wonju College of Medicine, Wonju, Ganwong 26460, Korea
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pratheek Bellur Marthanda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Lang Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Mattias Magnusson
- The National Board of Health and Welfare, Socialstyrelsen, 11259 Stockholm, Sweden
| | - Oleg Sysoev
- Department of Computer and Information Science, Linköping University, 58183 Linköping, Sweden
| | - Huan Zhang
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden
| | - Yelin Zhao
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Christopher Sjöwall
- Biomedical and Clinical Sciences, Division of Inflammation and Infection/Rheumatology, Linköping University, 58183 Linköping, Sweden
| | - Danuta Gawel
- Mavatar, Inc, Vasagatan, 11120 Stockholm, Sweden
| | - Hui Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, China
| | - Mikael Benson
- Department of Pediatrics, Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden; Medical Digital Twin Research Group, Division of Ear, Nose and Throat Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17165 Stockholm, Sweden.
| |
Collapse
|
18
|
Ghaffarinia A, Ayaydin F, Póliska S, Manczinger M, Bolla BS, Flink LB, Balogh F, Veréb Z, Bozó R, Szabó K, Bata-Csörgő Z, Kemény L. Psoriatic Resolved Skin Epidermal Keratinocytes Retain Disease-Residual Transcriptomic and Epigenomic Profiles. Int J Mol Sci 2023; 24:ijms24054556. [PMID: 36901987 PMCID: PMC10002496 DOI: 10.3390/ijms24054556] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
The disease-residual transcriptomic profile (DRTP) within psoriatic healed/resolved skin and epidermal tissue-resident memory T (TRM) cells have been proposed to be crucial for the recurrence of old lesions. However, it is unclear whether epidermal keratinocytes are involved in disease recurrence. There is increasing evidence regarding the importance of epigenetic mechanisms in the pathogenesis of psoriasis. Nonetheless, the epigenetic changes that contribute to the recurrence of psoriasis remain unknown. The aim of this study was to elucidate the role of keratinocytes in psoriasis relapse. The epigenetic marks 5-methylcytosine (5-mC) and 5-hydroxymethylcytosine (5-hmC) were visualized using immunofluorescence staining, and RNA sequencing was performed on paired never-lesional and resolved epidermal and dermal compartments of skin from psoriasis patients. We observed diminished 5-mC and 5-hmC amounts and decreased mRNA expression of the ten-eleven translocation (TET) 3 enzyme in the resolved epidermis. SAMHD1, C10orf99, and AKR1B10: the highly dysregulated genes in resolved epidermis are known to be associated with pathogenesis of psoriasis, and the DRTP was enriched in WNT, TNF, and mTOR signaling pathways. Our results suggest that epigenetic changes detected in epidermal keratinocytes of resolved skin may be responsible for the DRTP in the same regions. Thus, the DRTP of keratinocytes may contribute to site-specific local relapse.
Collapse
Affiliation(s)
- Ameneh Ghaffarinia
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
| | - Ferhan Ayaydin
- HCEMM-USZ, Functional Cell Biology and Immunology, Advanced Core Facility, H-6728 Szeged, Hungary
- Laboratory of Cellular Imaging, Biological Research Centre, Eötvös Loránd Research Network, H-6726 Szeged, Hungary
- Institute of Plant Biology, Biological Research Centre, H-6726 Szeged, Hungary
| | - Szilárd Póliska
- Genomic Medicine and Bioinformatics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Máté Manczinger
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- Systems Immunology Research Group, Institute of Biochemistry, Biological Research Centre, ELKH, H-6726 Szeged, Hungary
- HCEMM-Systems Immunology Research Group, H-6726 Szeged, Hungary
| | - Beáta Szilvia Bolla
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
| | - Lili Borbála Flink
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
| | - Fanni Balogh
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- ELKH-SZTE Dermatological Research Group, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory (HECRIN), Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
- Research Institute of Translational Biomedicine, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Renáta Bozó
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- ELKH-SZTE Dermatological Research Group, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Kornélia Szabó
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- ELKH-SZTE Dermatological Research Group, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Zsuzsanna Bata-Csörgő
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- ELKH-SZTE Dermatological Research Group, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
| | - Lajos Kemény
- HCEMM-USZ Skin Research Group, H-6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, H-6720 Szeged, Hungary
- ELKH-SZTE Dermatological Research Group, Department of Dermatology and Allergology, University of Szeged, H-6720 Szeged, Hungary
- Correspondence:
| |
Collapse
|
19
|
Mirghani H, Alharfy AAN, Alanazi AMM, Aljohani JKM, Aljohani RAA, Albalawi RHA, Aljohani RAA, Alqasmi Albalawi DM, Albalawi RHA, Mostafa MI. Diagnostic Test Accuracy of Genetic Tests in Diagnosing Psoriasis: A Systematic Review. Cureus 2022; 14:e31338. [PMID: 36514633 PMCID: PMC9741513 DOI: 10.7759/cureus.31338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2022] [Indexed: 11/12/2022] Open
Abstract
The pathogenesis of psoriasis involves the interaction of several environmental and genetic factors. Predicting the disease risk cannot depend on individual genetic alleles. Consequently, some studies have evaluated the use of genetic risk scores that combine several psoriasis susceptibility loci to increase the accuracy of predicting/diagnosing the disease. This meta-analysis summarizes the evidence regarding using genetic risk scores (GRS) in the diagnosis or prediction of psoriasis. A search of MEDLINE/PubMed, the Latin American Caribbean Health Sciences Literature (LILACS) database, Cochrane Library, Scopus, Web of Science, and ProQuest was conducted in July 2022. The primary objective was to record the area under the curve (AUC) for GRS of psoriasis. Secondary objectives included characteristics of studies and patients. The risk of bias (ROB) was assessed using the PROBAST tool. Five studies fulfilled the eligibility criteria of this review. None of the studies described the clinical criteria (reference standard) that were employed to diagnose psoriasis. The AUCs of the 11 GRS models ranged from 0.6029-0.8583 (median: 0.75). Marked heterogeneity was detected (Cochran Q: 1250.051, p < 0.001, and I2 index: 99.2%). So, pooling of the results of the included studies was not performed. The ROB was high for all studies and clinical application was not described. Genetic risk scores are promising tools for the prediction of psoriasis with fair to good accuracy. However, further research is required to identify the most accurate combination of loci and to validate the scores in variable ethnicities.
Collapse
Affiliation(s)
- Hyder Mirghani
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, Tabuk, SAU
| | | | | | | | | | | | | | | | | | - Mohamed I Mostafa
- Department of Anatomy, Faculty of Medicine, University of Tabuk, Tabuk, SAU
| |
Collapse
|
20
|
Fernández-Ruiz JC, Ochoa-González FDL, Zapata-Zúñiga M, Mondragon-Marín E, Lara-Ramírez EE, Ruíz-Carrillo JL, DelaCruz-Flores PA, Layseca-Espinosa E, Enciso-Moreno JA, Castañeda-Delgado JE. GPR15 expressed in T lymphocytes from RA patients is involved in leukocyte chemotaxis to the synovium. J Leukoc Biol 2022; 112:1209-1221. [PMID: 36164808 DOI: 10.1002/jlb.3ma0822-263rr] [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: 07/14/2021] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 12/24/2022] Open
Abstract
The rheumatoid arthritis (RA) inflammatory process occurs in the joints where immune cells are attracted into the synovium to promote remodeling and tissue damage. GPR15 is a G protein-coupled receptor (GPCR) located on chromosome 3 and has similarity in its sequence with chemokine receptors. Recent evidence indicates that GPR15 may be associated with modulation of the chronic inflammatory response. We evaluated the expression of GPR15 and GPR15L in blood and synovial tissue samples from RA patients, as well as to perform a functional migration assay in response to GPR15L. The expression of GPR15 and c10orf99/gpr15l mRNA was analyzed by RT-qPCR. Samples of synovial fluid and peripheral blood were analyzed for CD45+CD3+CD4+GPR15+ and CD45+CD3+CD8+GPR15+ T cell frequency comparing RA patients versus control subjects by flow cytometry. Migration assays were performed using PBMCs isolated from these individuals in response to the synthetic GPR15 ligand. Statistical analysis included Kruskal-Wallis test, T-test, or Mann-Whitney U test, according to data distribution. A higher expression in the mRNA for GPR15 was identified in early RA subjects. The frequencies of CD4+/CD8+ GPR15+ T lymphocytes are higher in RA patients comparing with healthy subjects. Also, the frequency CD4+/CD8+ GPR15+ T lymphocytes are higher in synovial fluid of established RA patients comparing with OA patients. GPR15 and GPR15L are present in the synovial tissue of RA patients and GPR15L promotes migration of PBMCs from RA patients and healthy subjects. Our results suggest that GPR15/GPR15L have a pathogenic role in RA and their antagonizing could be a therapeutic approach in RA.
Collapse
Affiliation(s)
- Julio Cesar Fernández-Ruiz
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México.,Centro de Investigación en Ciencias de la Salud y Biomedicina, Univerisidad Autónoma de San Luis Potosí, San Luis Potosí, San Luis Potosí, México
| | - Fátima de Lourdes Ochoa-González
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México.,Doctorado en ciencias básicas, Universidad Autónoma de Zacatecas, Zacatecas, Zacatecas, México.,Área de Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas, Zacatecas, México
| | - Martín Zapata-Zúñiga
- Hospital Rural No. 51 IMSS Bienestar, Villanueva, Zacatecas, México.,Unidad Académica de Medicina Humana y Ciencias de la Salud, Universidad Autónoma de Zacatecas, Zacatecas, Zacatecas, México
| | - Eduardo Mondragon-Marín
- Unidad de traumatología y ortopedia, Hospital general del Instituto Mexicano del Seguro Social Zacatecas "Emilio Varela Luján", Zacatecas, Zacatecas, México
| | - Edgar E Lara-Ramírez
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México
| | - Jose Luis Ruíz-Carrillo
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México.,Centro de Investigación en Ciencias de la Salud y Biomedicina, Univerisidad Autónoma de San Luis Potosí, San Luis Potosí, San Luis Potosí, México
| | - Paola Amayrani DelaCruz-Flores
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México
| | - Esther Layseca-Espinosa
- Centro de Investigación en Ciencias de la Salud y Biomedicina, Univerisidad Autónoma de San Luis Potosí, San Luis Potosí, San Luis Potosí, México
| | - José Antonio Enciso-Moreno
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México.,Maestría en química clínica diagnóstica, Facultad de Química, Universidad Autónoma de Querétaro, Santiago de Queretáro, Querétaro, México
| | - Julio Enrique Castañeda-Delgado
- Unidad de Investigación Biomédica de Zacatecas, Instituto Mexicano del Seguro Social (IMSS), Zacatecas, Zacatecas, México.,Cátedras CONACYT, Consejo Nacional de Ciencia y Tecnología, Ciudad de México, México
| |
Collapse
|
21
|
Deng M, Su Y, Wu R, Li S, Zhu Y, Tang G, Shi X, Zhou T, Zhao M, Lu Q. DNA methylation markers in peripheral blood for psoriatic arthritis. J Dermatol Sci 2022; 108:39-47. [DOI: 10.1016/j.jdermsci.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 10/25/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022]
|
22
|
Shi S, Pan X, Zhang L, Wang X, Zhuang Y, Lin X, Shi S, Zheng J, Lin W. An application based on bioinformatics and machine learning for risk prediction of sepsis at first clinical presentation using transcriptomic data. Front Genet 2022; 13:979529. [PMID: 36159979 PMCID: PMC9490444 DOI: 10.3389/fgene.2022.979529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Linking genotypic changes to phenotypic traits based on machine learning methods has various challenges. In this study, we developed a workflow based on bioinformatics and machine learning methods using transcriptomic data for sepsis obtained at the first clinical presentation for predicting the risk of sepsis. By combining bioinformatics with machine learning methods, we have attempted to overcome current challenges in predicting disease risk using transcriptomic data. Methods: High-throughput sequencing transcriptomic data processing and gene annotation were performed using R software. Machine learning models were constructed, and model performance was evaluated by machine learning methods in Python. The models were visualized and interpreted using the Shapley Additive explanation (SHAP) method. Results: Based on the preset parameters and using recursive feature elimination implemented via machine learning, the top 10 optimal genes were screened for the establishment of the machine learning models. In a comparison of model performance, CatBoost was selected as the optimal model. We explored the significance of each gene in the model and the interaction between each gene through SHAP analysis. Conclusion: The combination of CatBoost and SHAP may serve as the best-performing machine learning model for predicting transcriptomic and sepsis risks. The workflow outlined may provide a new approach and direction in exploring the mechanisms associated with genes and sepsis risk.
Collapse
Affiliation(s)
- Songchang Shi
- Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital South Branch, Fujian Provincial Jinshan Hospital, Fujian Provincial Hospital, Fuzhou, China
| | - Xiaobin Pan
- Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital South Branch, Fujian Provincial Jinshan Hospital, Fujian Provincial Hospital, Fuzhou, China
| | - Lihui Zhang
- Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital South Branch, Fujian Provincial Jinshan Hospital, Fujian Provincial Hospital, Fuzhou, China
| | - Xincai Wang
- Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital South Branch, Fujian Provincial Jinshan Hospital, Fujian Provincial Hospital, Fuzhou, China
| | - Yingfeng Zhuang
- Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital South Branch, Fujian Provincial Jinshan Hospital, Fujian Provincial Hospital, Fuzhou, China
| | - Xingsheng Lin
- Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital South Branch, Fujian Provincial Jinshan Hospital, Fujian Provincial Hospital, Fuzhou, China
| | - Songjing Shi
- Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Jianzhang Zheng
- Department of Orthopedics, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Wei Lin
- Department of Endocrinology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| |
Collapse
|
23
|
Song J, Li Z, Yao G, Wei S, Li L, Wu H. Framework for feature selection of predicting the diagnosis and prognosis of necrotizing enterocolitis. PLoS One 2022; 17:e0273383. [PMID: 35984833 PMCID: PMC9390903 DOI: 10.1371/journal.pone.0273383] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/08/2022] [Indexed: 11/18/2022] Open
Abstract
Neonatal necrotizing enterocolitis (NEC) occurs worldwide and is a major source of neonatal morbidity and mortality. Researchers have developed many methods for predicting NEC diagnosis and prognosis. However, most people use statistical methods to select features, which may ignore the correlation between features. In addition, because they consider a small dimension of characteristics, they neglect some laboratory parameters such as white blood cell count, lymphocyte percentage, and mean platelet volume, which could be potentially influential factors affecting the diagnosis and prognosis of NEC. To address these issues, we include more perinatal, clinical, and laboratory information, including anemia—red blood cell transfusion and feeding strategies, and propose a ridge regression and Q-learning strategy based bee swarm optimization (RQBSO) metaheuristic algorithm for predicting NEC diagnosis and prognosis. Finally, a linear support vector machine (linear SVM), which specializes in classifying high-dimensional features, is used as a classifier. In the NEC diagnostic prediction experiment, the area under the receiver operating characteristic curve (AUROC) of dataset 1 (feeding intolerance + NEC) reaches 94.23%. In the NEC prognostic prediction experiment, the AUROC of dataset 2 (medical NEC + surgical NEC) reaches 91.88%. Additionally, the classification accuracy of the RQBSO algorithm on the NEC dataset is higher than the other feature selection algorithms. Thus, the proposed approach has the potential to identify predictors that contribute to the diagnosis of NEC and stratification of disease severity in a clinical setting.
Collapse
Affiliation(s)
- Jianfei Song
- College of Communication Engineering, Jilin University, Changchun, Jilin, PR China
| | - Zhenyu Li
- Department of Neonatology, Jilin University First Hospital, Changchun, Jilin, PR China
| | - Guijin Yao
- College of Communication Engineering, Jilin University, Changchun, Jilin, PR China
| | - Songping Wei
- College of Communication Engineering, Jilin University, Changchun, Jilin, PR China
| | - Ling Li
- College of Communication Engineering, Jilin University, Changchun, Jilin, PR China
- * E-mail: (LL); (HW)
| | - Hui Wu
- Department of Neonatology, Jilin University First Hospital, Changchun, Jilin, PR China
- * E-mail: (LL); (HW)
| |
Collapse
|
24
|
Abstract
AIMS We aimed to develop a gene signature that predicts the occurrence of postmenopausal osteoporosis (PMOP) by studying its genetic mechanism. METHODS Five datasets were obtained from the Gene Expression Omnibus database. Unsupervised consensus cluster analysis was used to determine new PMOP subtypes. To determine the central genes and the core modules related to PMOP, the weighted gene co-expression network analysis (WCGNA) was applied. Gene Ontology enrichment analysis was used to explore the biological processes underlying key genes. Logistic regression univariate analysis was used to screen for statistically significant variables. Two algorithms were used to select important PMOP-related genes. A logistic regression model was used to construct the PMOP-related gene profile. The receiver operating characteristic area under the curve, Harrell's concordance index, a calibration chart, and decision curve analysis were used to characterize PMOP-related genes. Then, quantitative real-time polymerase chain reaction (qRT-PCR) was used to verify the expression of the PMOP-related genes in the gene signature. RESULTS We identified three PMOP-related subtypes and four core modules. The muscle system process, muscle contraction, and actin filament-based movement were more active in the hub genes. We obtained five feature genes related to PMOP. Our analysis verified that the gene signature had good predictive power and applicability. The outcomes of the GSE56815 cohort were found to be consistent with the results of the earlier studies. qRT-PCR results showed that RAB2A and FYCO1 were amplified in clinical samples. CONCLUSION The PMOP-related gene signature we developed and verified can accurately predict the risk of PMOP in patients. These results can elucidate the molecular mechanism of RAB2A and FYCO1 underlying PMOP, and yield new and improved treatment strategies, ultimately helping PMOP monitoring.Cite this article: Bone Joint Res 2022;11(8):548-560.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| | - Maowei Yang
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| | - Yue Zhu
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
25
|
Tian Y, Lu Y, Cao Y, Dang C, Wang N, Tian K, Luo Q, Guo E, Luo S, Wang L, Li Q. Identification of diagnostic signatures associated with immune infiltration in Alzheimer’s disease by integrating bioinformatic analysis and machine-learning strategies. Front Aging Neurosci 2022; 14:919614. [PMID: 35966794 PMCID: PMC9372364 DOI: 10.3389/fnagi.2022.919614] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/11/2022] [Indexed: 12/04/2022] Open
Abstract
Objective As a chronic neurodegenerative disorder, Alzheimer’s disease (AD) is the most common form of progressive dementia. The purpose of this study was to identify diagnostic signatures of AD and the effect of immune cell infiltration in this pathology. Methods The expression profiles of GSE109887, GSE122063, GSE28146, and GSE1297 were downloaded from the Gene Expression Omnibus (GEO) database to obtain differentially expressed genes (DEGs) between AD and control brain samples. Functional enrichment analysis was performed to reveal AD-associated biological functions and key pathways. Besides, we applied the Least Absolute Shrinkage Selection Operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE) analysis to screen potential diagnostic feature genes in AD, which were further tested in AD brains of the validation cohort (GSE5281). The discriminatory ability was then assessed by the area under the receiver operating characteristic curves (AUC). Finally, the CIBERSORT algorithm and immune cell infiltration analysis were employed to assess the inflammatory state of AD. Results A total of 49 DEGs were identified. The functional enrichment analysis revealed that leukocyte transendothelial migration, cytokine receptor interaction, and JAK-STAT signaling pathway were enriched in the AD group. MAF basic leucine zipper transcription factor F (MAFF), ADCYAP1, and ZFP36L1 were identified as the diagnostic biomarkers of AD with high discriminatory ability (AUC = 0.850) and validated in AD brains (AUC = 0.935). As indicated from the immune cell infiltration analysis, naive B cells, plasma cells, activated/resting NK cells, M0 macrophages, M1 macrophages, resting CD4+ T memory cells, resting mast cells, memory B cells, and resting/activated dendritic cells may participate in the development of AD. Additionally, all diagnostic signatures presented different degrees of correlation with different infiltrating immune cells. Conclusion MAFF, ADCYAP1, and ZFP36L1 may become new candidate biomarkers of AD, which were closely related to the pathogenesis of AD. Moreover, the immune cells mentioned above may play crucial roles in disease occurrence and progression.
Collapse
Affiliation(s)
- Yu Tian
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Gerontology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yaoheng Lu
- Department of General Surgery, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Chengdu, China
| | - Yuze Cao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Chun Dang
- West China Medical Publishers, West China Hospital, Sichuan University, Chengdu, China
| | - Na Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kuo Tian
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiqi Luo
- Department of Gerontology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Erliang Guo
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shanshun Luo
- Department of Gerontology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- Shanshun Luo,
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Lihua Wang,
| | - Qian Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Qian Li,
| |
Collapse
|
26
|
Rezaee K, Jeon G, Khosravi MR, Attar HH, Sabzevari A. Deep learning‐based microarray cancer classification and ensemble gene selection approach. IET Syst Biol 2022; 16:120-131. [PMID: 35790076 PMCID: PMC9290776 DOI: 10.1049/syb2.12044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/04/2022] [Accepted: 05/31/2022] [Indexed: 12/19/2022] Open
Abstract
Malignancies and diseases of various genetic origins can be diagnosed and classified with microarray data. There are many obstacles to overcome due to the large size of the gene and the small number of samples in the microarray. A combination strategy for gene expression in a variety of diseases is described in this paper, consisting of two steps: identifying the most effective genes via soft ensembling and classifying them with a novel deep neural network. The feature selection approach combines three strategies to select wrapper genes and rank them according to the k‐nearest neighbour algorithm, resulting in a very generalisable model with low error levels. Using soft ensembling, the most effective subsets of genes were identified from three microarray datasets of diffuse large cell lymphoma, leukaemia, and prostate cancer. A stacked deep neural network was used to classify all three datasets, achieving an average accuracy of 97.51%, 99.6%, and 96.34%, respectively. In addition, two previously unreported datasets from small, round blue cell tumors (SRBCTs)and multiple sclerosis‐related brain tissue lesions were examined to show the generalisability of the model method.
Collapse
Affiliation(s)
- Khosro Rezaee
- Department of Biomedical Engineering Meybod University Meybod Iran
| | - Gwanggil Jeon
- Department of Embedded Systems Engineering College of Information Technology Incheon National University Incheon Korea
| | | | - Hani H. Attar
- Department of Energy Engineering Zarqa University Zarqa Jordan
| | | |
Collapse
|
27
|
Ahmed H, Soliman H, Elmogy M. Early detection of Alzheimer's disease using single nucleotide polymorphisms analysis based on gradient boosting tree. Comput Biol Med 2022; 146:105622. [PMID: 35751201 DOI: 10.1016/j.compbiomed.2022.105622] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD) is a degenerative disorder that attacks nerve cells in the brain. AD leads to memory loss and cognitive & intellectual impairments that can influence social activities and decision-making. The most common type of human genetic variation is single nucleotide polymorphisms (SNPs). SNPs are beneficial markers of complex gene-disease. Many common and serious diseases, such as AD, have associated SNPs. Detection of SNP biomarkers linked with AD could help in the early prediction and diagnosis of this disease. The main objective of this paper is to predict and diagnose AD based on SNPs biomarkers with high classification accuracy in the early stages. One of the most concerning problems is the high number of features. Thus, the paper proposes a comprehensive framework for early AD detection and detecting the most significant genes based on SNPs analysis. Usage of machine learning (ML) techniques to identify new biomarkers of AD is also suggested. In the proposed system, two feature selection techniques are separately checked: the information gain filter and Boruta wrapper. The two feature selection techniques were used to select the most significant genes related to AD in this system. Filter methods measure the relevance of features by their correlation with dependent variables, while wrapper methods measure the usefulness of a subset of features by training a model on it. Gradient boosting tree (GBT) has been applied on all AD genetic data of neuroimaging initiative phase 1 (ADNI-1) and Whole-Genome Sequencing (WGS) datasets by using two feature selection techniques. In the whole-genome approach ADNI-1, results revealed that the GBT learning algorithm scored an overall accuracy of 99.06% in the case of using Boruta feature selection. Using information gain feature selection, the proposed system achieved an average accuracy of 94.87%. The results show that the proposed system is preferable for the early detection of AD. Also, the results revealed that the Boruta wrapper feature selection is superior to the information gain filter technique.
Collapse
Affiliation(s)
- Hala Ahmed
- Information Technology Dept., Faculty of Computers and Information, Mansoura University, Mansoura, P.O.35516, Egypt
| | - Hassan Soliman
- Information Technology Dept., Faculty of Computers and Information, Mansoura University, Mansoura, P.O.35516, Egypt
| | - Mohammed Elmogy
- Information Technology Dept., Faculty of Computers and Information, Mansoura University, Mansoura, P.O.35516, Egypt.
| |
Collapse
|
28
|
Tseng PY, Hoon MA. GPR15L is an epithelial inflammation-derived pruritogen. SCIENCE ADVANCES 2022; 8:eabm7342. [PMID: 35704588 PMCID: PMC9200282 DOI: 10.1126/sciadv.abm7342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 05/02/2022] [Indexed: 05/09/2023]
Abstract
Itch is an unpleasant sensation that often accompanies chronic dermatological conditions. Although many of the itch receptors and the neural pathways underlying this sensation are known, the identity of endogenous ligands is still not fully appreciated. Using an unbiased bioinformatic approach, we identified GPR15L as a candidate pruritogen whose expression is robustly up-regulated in psoriasis and atopic dermatitis. Although GPR15L was previously shown to be a cognate ligand of the receptor GPR15, expressed in dermal T cells, here we show that it also contributes to pruritogenesis by activating Mas-related G protein-coupled receptors (MRGPRs). GPR15L can selectively stimulate mouse dorsal root ganglion neurons that express Mrgpra3 and evokes intense itch responses. GPR15L causes mast cell degranulation through stimulation of MRGPRX2 and Mrgprb2. Genetic disruption of GPR15L expression attenuates scratch responses in a mouse model of psoriasis. Our study reveals unrecognized features of GRP15L, showing that it is a potent itch-inducing agent.
Collapse
Affiliation(s)
- Pang-Yen Tseng
- Molecular Genetics Section, National Institute of Dental and Craniofacial Research/NIH, 35 Convent Drive, Bethesda, MD 20892, USA
| | | |
Collapse
|
29
|
Wang Q, Duan M, Fan Y, Liu S, Ren Y, Huang L, Zhou F. Transforming OMIC features for classification using Siamese convolutional networks. J Bioinform Comput Biol 2022; 20:2250013. [DOI: 10.1142/s0219720022500135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
30
|
Dainichi T, Nakano Y, Doi H, Nakamizo S, Nakajima S, Matsumoto R, Farkas T, Wong PM, Narang V, Moreno Traspas R, Kawakami E, Guttman-Yassky E, Dreesen O, Litman T, Reversade B, Kabashima K. C10orf99/GPR15L Regulates Proinflammatory Response of Keratinocytes and Barrier Formation of the Skin. Front Immunol 2022; 13:825032. [PMID: 35273606 PMCID: PMC8902463 DOI: 10.3389/fimmu.2022.825032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/26/2022] [Indexed: 12/13/2022] Open
Abstract
The epidermis, outermost layer of the skin, forms a barrier and is involved in innate and adaptive immunity in an organism. Keratinocytes participate in all these three protective processes. However, a regulator of keratinocyte protective responses against external dangers and stresses remains elusive. We found that upregulation of the orphan gene 2610528A11Rik was a common factor in the skin of mice with several types of inflammation. In the human epidermis, peptide expression of G protein-coupled receptor 15 ligand (GPR15L), encoded by the human ortholog C10orf99, was highly induced in the lesional skin of patients with atopic dermatitis or psoriasis. C10orf99 gene transfection into normal human epidermal keratinocytes (NHEKs) induced the expression of inflammatory mediators and reduced the expression of barrier-related genes. Gene ontology analyses showed its association with translation, mitogen-activated protein kinase (MAPK), mitochondria, and lipid metabolism. Treatment with GPR15L reduced the expression levels of filaggrin and loricrin in human keratinocyte 3D cultures. Instead, their expression levels in mouse primary cultured keratinocytes did not show significant differences between the wild-type and 2610528A11Rik deficient keratinocytes. Lipopolysaccharide-induced expression of Il1b and Il6 was less in 2610528A11Rik deficient mouse keratinocytes than in wild-type, and imiquimod-induced psoriatic dermatitis was blunted in 2610528A11Rik deficient mice. Furthermore, repetitive subcutaneous injection of GPR15L in mouse ears induced skin inflammation in a dose-dependent manner. These results suggest that C10orf99/GPR15L is a primary inducible regulator that reduces the barrier formation and induces the inflammatory response of keratinocytes.
Collapse
Affiliation(s)
- Teruki Dainichi
- Department of Dermatology, Faculty of Medicine, Kagawa University, Miki-cho, Japan
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuri Nakano
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiromi Doi
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Nakamizo
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Agency for Science, Technology and Research (A*STAR) Skin Research Laboratories (A*SRL), A*STAR, Biopolis, Singapore, Singapore
| | - Saeko Nakajima
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Drug Discovery for Inflammatory Skin Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Reiko Matsumoto
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Thomas Farkas
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Pui Mun Wong
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Vipin Narang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Ricardo Moreno Traspas
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Eiryo Kawakami
- Advanced Data Science Project (ADSP), RIKEN, Yokohama, Japan
- Artificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Oliver Dreesen
- Agency for Science, Technology and Research (A*STAR) Skin Research Laboratories (A*SRL), A*STAR, Biopolis, Singapore, Singapore
| | - Thomas Litman
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Bruno Reversade
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore, Singapore
| | - Kenji Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Agency for Science, Technology and Research (A*STAR) Skin Research Laboratories (A*SRL), A*STAR, Biopolis, Singapore, Singapore
| |
Collapse
|
31
|
Lin S, Lin Y, Wu K, Wang Y, Feng Z, Duan M, Liu S, Fan Y, Huang L, Zhou F. FeCO3, constructing the network biomarkers using the inter-feature correlation coefficients and its application in detecting high-order breast cancer biomarkers. Curr Bioinform 2022. [DOI: 10.2174/1574893617666220124123303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aims:
This study aims to formulate the inter-feature correlation as the engineered features.
Background:
Modern biotechnologies tend to generate a huge number of characteristics of a sample, while an OMIC dataset usually has a few dozens or hundreds of samples due to the high costs of generating the OMIC data. So many bio-OMIC studies assumed the inter-feature independence and selected a feature with a high phenotype-association.
Objective:
However, many features are closely associated with each other due to their physical or functional interactions, which may be utilized as a new view of features.
Method:
This study proposed a feature engineering algorithm based on the correlation coefficients (FeCO3) by utilizing the correlations between a given sample and a few reference samples. A comprehensive evaluation was carried out for the proposed FeCO3 network features using 24 bio-OMIC datasets.
Result:
The experimental data suggested that the newly calculated FeCO3 network features tended to achieve better classification performances than the original features, using the same popular feature selection and classification algorithms. The FeCO3 network features were also consistently supported by the literature. FeCO3 was utilized to investigate the high-order engineered biomarkers of breast cancer, and detected the PBX2 gene (Pre-B-Cell Leukemia Transcription Factor 2) as one of the candidate breast cancer biomarkers. Although the two methylated residues cg14851325 (Pvalue=8.06e-2) and cg16602460 (Pvalue=1.19e-1) within PBX2 did not have statistically significant association with breast cancers, the high-order inter-feature correlations showed a significant association with breast cancers.
Conclusion:
The proposed FeCO3 network features calculated the high-order inter-feature correlations as novel features, and may facilitate the investigations of complex diseases from this new perspective. The source code is available in FigShare at 10.6084/m9.figshare.13550051 or the web site http://www.healthinformaticslab.org/supp/ .
Collapse
Affiliation(s)
- Shenggeng Lin
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuqi Lin
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Kexin Wu
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Yueying Wang
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Zixuan Feng
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Meiyu Duan
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Shuai Liu
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Yusi Fan
- College of Software, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Lan Huang
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| | - Fengfeng Zhou
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
32
|
Martorell A, Martin-Gorgojo A, Ríos-Viñuela E, Rueda-Carnero J, Alfageme F, Taberner R. [Translated article] Artificial intelligence in dermatology: A threat or an opportunity? ACTAS DERMO-SIFILIOGRAFICAS 2022. [DOI: 10.1016/j.ad.2021.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
33
|
Martorell A, Martin-Gorgojo A, Ríos-Viñuela E, Rueda-Carnero J, Alfageme F, Taberner R. Inteligencia artificial en dermatología: ¿amenaza u oportunidad? ACTAS DERMO-SIFILIOGRAFICAS 2022; 113:30-46. [DOI: 10.1016/j.ad.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/18/2021] [Indexed: 11/25/2022] Open
|
34
|
Martorell A, Martin-Gorgojo A, Ríos-Viñuela E, Rueda-Carnero J, Alfageme F, Taberner R. Artificial intelligence in dermatology: A threat or an opportunity? ACTAS DERMO-SIFILIOGRAFICAS 2021. [DOI: 10.1016/j.adengl.2021.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
35
|
Guo Y, Yin J, Dai Y, Guan Y, Chen P, Chen Y, Huang C, Lu YJ, Zhang L, Song D. A Novel CpG Methylation Risk Indicator for Predicting Prognosis in Bladder Cancer. Front Cell Dev Biol 2021; 9:642650. [PMID: 34540821 PMCID: PMC8440888 DOI: 10.3389/fcell.2021.642650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 08/05/2021] [Indexed: 01/15/2023] Open
Abstract
Purpose Bladder cancer (BLCA) is one of the most common cancers worldwide. In a large proportion of BLCA patients, disease recurs and/or progress after resection, which remains a major clinical issue in BLCA management. Therefore, it is vital to identify prognostic biomarkers for treatment stratification. We investigated the efficiency of CpG methylation for the potential to be a prognostic biomarker for patients with BLCA. Patients and Methods Overall, 357 BLCA patients from The Cancer Genome Atlas (TCGA) were randomly separated into the training and internal validation cohorts. Least absolute shrinkage and selector operation (LASSO) and support vector machine-recursive feature elimination (SVM-RFE) were used to select candidate CpGs and build the methylation risk score model, which was validated for its prognostic value in the validation cohort by Kaplan–Meier analysis. Hazard curves were generated to reveal the risk nodes throughout the follow-up. Gene Set Enrichment Analysis (GSEA) was used to reveal the potential biological pathways associated with the methylation model. Quantitative real-time polymerase chain reaction (PCR) and western blotting were performed to verify the expression level of the methylated genes. Results After incorporating the CpGs obtained by the two algorithms, CpG methylation of eight genes corresponding to TNFAIP8L3, KRTDAP, APC, ZC3H3, COL9A2, SLCO4A1, POU3F3, and ADARB2 were prominent candidate predictors in establishing a methylation risk score for BLCA (MRSB), which was used to divide the patients into high- and low-risk progression groups (p < 0.001). The effectiveness of the MRSB was validated in the internal cohort (p < 0.001). In the MRSB high-risk group, the hazard curve exhibited an initial wide, high peak within 10 months after treatment, whereas some gentle peaks around 2 years were noted. Furthermore, a nomogram comprising MRSB, age, sex, and tumor clinical stage was developed to predict the individual progression risk, and it performed well. Survival analysis implicated the effectiveness of MRSB, which remains significant in all the subgroup analysis based on the clinical features. A functional analysis of MRSB and the corresponding genes revealed potential pathways affecting tumor progression. Validation of quantitative real-time PCR and western blotting revealed that TNFAIP8L3 was upregulated in the BLCA tissues. Conclusion We developed the MRSB, an eight-gene-based methylation signature, which has great potential to be used to predict the post-surgery progression risk of BLCA.
Collapse
Affiliation(s)
- Yufeng Guo
- Department of Urology, The First Affiliated Hospital & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jianjian Yin
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yuanheng Dai
- Department of Urology, The First Affiliated Hospital & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yudong Guan
- Department of Urology, The First Affiliated Hospital & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Pinjin Chen
- Department of Urology, The First Affiliated Hospital & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yongqiang Chen
- Department of Urology, The First Affiliated Hospital & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chenzheng Huang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yong-Jie Lu
- Department of Urology, The First Affiliated Hospital & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China.,Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Dongkui Song
- Department of Urology, The First Affiliated Hospital & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Tapak L, Afshar S, Afrasiabi M, Ghasemi MK, Alirezaei P. Application of Genetic Algorithm-Based Support Vector Machine in Identification of Gene Expression Signatures for Psoriasis Classification: A Hybrid Model. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5520710. [PMID: 34540995 PMCID: PMC8443357 DOI: 10.1155/2021/5520710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/23/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Psoriasis is a chronic autoimmune disease impairing significantly the quality of life of the patient. The diagnosis of the disease is done via a visual inspection of the lesional skin by dermatologists. Classification of psoriasis using gene expression is an important issue for the early and effective treatment of the disease. Therefore, gene expression data and selection of suitable gene signatures are effective sources of information. METHODS We aimed to develop a hybrid classifier for the diagnosis of psoriasis based on two machine learning models of the genetic algorithm and support vector machine (SVM). The method also conducts gene signature selection. A publically available gene expression dataset was used to test the model. RESULTS A number of 181 probe sets were selected among the original 54,675 probes using the hybrid model with a prediction accuracy of 100% over the test set. A number of 10 hub genes were identified using the protein-protein interaction network. Nine out of 10 identified genes were found in significant modules. CONCLUSIONS The results showed that the genetic algorithm improved the SVM classifier performance significantly implying the ability of the proposed model in terms of detecting relevant gene expression signatures as the best features.
Collapse
Affiliation(s)
- Leili Tapak
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
- Modeling of Noncommunicable Diseases Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saeid Afshar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mohammad Kazem Ghasemi
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Pedram Alirezaei
- Department of Dermatology, Psoriasis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
37
|
Nanda MA, Seminar KB, Maddu A, Nandika D. Identifying relevant features of termite signals applied in termite detection system. ECOL INFORM 2021. [DOI: 10.1016/j.ecoinf.2021.101391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
38
|
Manohar M, Dunham D, Gupta S, Yan Z, Zhang W, Minnicozzi S, Kirkey M, Bunning B, Chowdhury RR, Galli SJ, Boyd SD, Kost LE, Chinthrajah RS, Desai M, Oettgen HC, Maecker HT, Yu W, DeKruyff RH, Andorf S, Nadeau KC. Immune changes beyond Th2 pathways during rapid multifood immunotherapy enabled with omalizumab. Allergy 2021; 76:2809-2826. [PMID: 33782956 PMCID: PMC8609920 DOI: 10.1111/all.14833] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/03/2021] [Accepted: 02/15/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Multifood oral immunotherapy (mOIT) with adjunctive anti-IgE (omalizumab, XOLAIR® ) treatment affords safe, effective, and rapid desensitization to multiple foods, although the specific immune mechanisms mediating this desensitization remain to be fully elucidated. METHODS Participants in our phase 2 mOIT trial (NCT02643862) received omalizumab from baseline to week 16 and mOIT from week 8 to week 36. We compared the immune profile of PBMCs and plasma taken at baseline, week 8, and week 36 using high-dimensional mass cytometry, component-resolved diagnostics, the indirect basophil activation test, and Luminex. RESULTS We found (i) decreased frequency of IL-4+ peanut-reactive CD4+ T cells and a marked downregulation of GPR15 expression and CXCR3 frequency among γδ and CD8+ T-cell subsets at week 8 during the initial, omalizumab-alone induction phase; (ii) significant upregulation of the skin-homing receptor CCR4 in peanut-reactive CD4+ T and Th2 effector memory (EM) cells and of cutaneous lymphocyte-associated antigen (CLA) in peanut-reactive CD8+ T and CD8+ EM cells; (iii) downregulation of CD86 expression among antigen-presenting cell subsets; and (iv) reduction in pro-inflammatory cytokines, notably IL-17, at week 36 post-OIT. We also observed significant attenuation of the Th2 phenotype post-OIT, defined by downregulation of IL-4 peanut-reactive T cells and OX40 in Th2EM cells, increased allergen component-specific IgG4/IgE ratio, and decreased allergen-driven activation of indirectly sensitized basophils. CONCLUSIONS This exploratory study provides novel comprehensive insight into the immune underpinnings of desensitization through omalizumab-facilitated mOIT. Moreover, this study provides encouraging results to support the complex immune changes that can be induced by OIT.
Collapse
Affiliation(s)
- Monali Manohar
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
| | - Diane Dunham
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
| | - Sheena Gupta
- Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA
| | - Zheng Yan
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
| | - Wenming Zhang
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
| | - Samantha Minnicozzi
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA; Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Matthew Kirkey
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
| | - Bryan Bunning
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
| | | | - Stephen J. Galli
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
- Department of Microbiology and Immunology, Stanford, CA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Scott D. Boyd
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | | | | | - Manisha Desai
- Department of Biomedical and Data Science, Stanford University, Stanford, CA
| | - Hans C. Oettgen
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Boston, MA; Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Holden T. Maecker
- Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA
- Department of Microbiology and Immunology, Stanford, CA
| | - Wong Yu
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
| | | | - Sandra Andorf
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, Divisions of Biomedical Informatics and Allergy & Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kari C. Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA
| |
Collapse
|
39
|
Peng C, Wu X, Yuan W, Zhang X, Zhang Y, Li Y. MGRFE: Multilayer Recursive Feature Elimination Based on an Embedded Genetic Algorithm for Cancer Classification. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:621-632. [PMID: 31180870 DOI: 10.1109/tcbb.2019.2921961] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Microarray gene expression data have become a topic of great interest for cancer classification and for further research in the field of bioinformatics. Nonetheless, due to the "large p, small n" paradigm of limited biosamples and high-dimensional data, gene selection is becoming a demanding task, which is aimed at selecting a minimal number of discriminatory genes associated closely with a phenotype. Feature or gene selection is still a challenging problem owing to its nondeterministic polynomial time complexity and thus most of the existing feature selection algorithms utilize heuristic rules. A multilayer recursive feature elimination method based on an embedded integer-coded genetic algorithm, MGRFE, is proposed here, which is aimed at selecting the gene combination with minimal size and maximal information. On the basis of 19 benchmark microarray datasets including multiclass and imbalanced datasets, MGRFE outperforms state-of-the-art feature selection algorithms with better cancer classification accuracy and a smaller selected gene number. MGRFE could be regarded as a promising feature selection method for high-dimensional datasets especially gene expression data. Moreover, the genes selected by MGRFE have close biological relevance to cancer phenotypes. The source code of our proposed algorithm and all the 19 datasets used in this paper are available at https://github.com/Pengeace/MGRFE-GaRFE.
Collapse
|
40
|
Han Y, Huang L, Zhou F. A dynamic recursive feature elimination framework (dRFE) to further refine a set of OMIC biomarkers. Bioinformatics 2021; 37:2183-2189. [PMID: 33515240 DOI: 10.1093/bioinformatics/btab055] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/23/2020] [Accepted: 01/25/2021] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION A feature selection algorithm may select the subset of features with the best associations with the class labels. The recursive feature elimination (RFE) is a heuristic feature screening framework and has been widely used to select the biological OMIC biomarkers. This study proposed a dynamic recursive feature elimination (dRFE) framework with more flexible feature elimination operations. The proposed dRFE was comprehensively compared with 11 existing feature selection algorithms and five classifiers on the eight difficult transcriptome datasets from a previous study, the ten newly collected transcriptome datasets and the five methylome datasets. RESULTS The experimental data suggested that the regular RFE framework did not perform well, and dRFE outperformed the existing feature selection algorithms in most cases. The dRFE-detected features achieved Acc=1.0000 for the two methylome datasets GSE53045 and GSE66695. The best prediction accuracies of the dRFE-detected features were 0.9259, 0.9424, and 0.8601 for the other three methylome datasets GSE74845, GSE103186, and GSE80970, respectively. Four transcriptome datasets received Acc=1.0000 using the dRFE-detected features, and the prediction accuracies for the other six newly collected transcriptome datasets were between 0.6301 and 0.9917. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yuanyuan Han
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin, China, 130012
| | - Lan Huang
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin, China, 130012
| | - Fengfeng Zhou
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin, China, 130012
| |
Collapse
|
41
|
Choudhary S, Pradhan D, Khan NS, Singh H, Thomas G, Jain AK. Decoding Psoriasis: Integrated Bioinformatics Approach to Understand Hub Genes and Involved Pathways. Curr Pharm Des 2021; 26:3619-3630. [PMID: 32160841 DOI: 10.2174/1381612826666200311130133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/22/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Psoriasis is a chronic immune mediated skin disorder with global prevalence of 0.2- 11.4%. Despite rare mortality, the severity of the disease could be understood by the accompanying comorbidities, that has even led to psychological problems among several patients. The cause and the disease mechanism still remain elusive. OBJECTIVE To identify potential therapeutic targets and affecting pathways for better insight of the disease pathogenesis. METHOD The gene expression profile GSE13355 and GSE14905 were retrieved from NCBI, Gene Expression Omnibus database. The GEO profiles were integrated and the DEGs of lesional and non-lesional psoriasis skin were identified using the affy package in R software. The Kyoto Encyclopaedia of Genes and Genomes pathways of the DEGs were analyzed using clusterProfiler. Cytoscape, V3.7.1 was utilized to construct protein interaction network and analyze the interactome map of candidate proteins encoded in DEGs. Functionally relevant clusters were detected through Cytohubba and MCODE. RESULTS A total of 1013 genes were differentially expressed in lesional skin of which 557 were upregulated and 456 were downregulated. Seven dysregulated genes were extracted in non-lesional skin. The disease gene network of these DEGs revealed 75 newly identified differentially expressed gene that might have a role in development and progression of the disease. GO analysis revealed keratinocyte differentiation and positive regulation of cytokine production to be the most enriched biological process and molecular function. Cytokines -cytokine receptor was the most enriched pathways. Among 1013 identified DEGs in lesional group, 36 DEGs were found to have altered genetic signature including IL1B and STAT3 which are also reported as hub genes. CCNB1, CCNA2, CDK1, IL1B, CXCL8, MKI 67, ESR1, UBE2C, STAT1 and STAT3 were top 10 hub gene. CONCLUSION The hub genes, genomic altered DEGs and other newly identified differentially dysregulated genes would improve our understanding of psoriasis pathogenesis, moreover, the hub genes could be explored as potential therapeutic targets for psoriasis.
Collapse
Affiliation(s)
- Saumya Choudhary
- Department of Molecular and Cellular Engineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj (Allahabad), India
| | - Dibyabhaba Pradhan
- ICMR-AIIMS Computational Genomics Centre (ISRM) Division- Indian Council of Medical Research, New Delhi, India
| | - Noor S Khan
- Biomedical Informatics Centre, National Institute of Pathology - Indian Council of Medical Research, New Delhi, India
| | - Harpreet Singh
- ICMR-AIIMS Computational Genomics Centre (ISRM) Division- Indian Council of Medical Research, New Delhi, India
| | - George Thomas
- Department of Molecular and Cellular Engineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj (Allahabad), India
| | - Arun K Jain
- Biomedical Informatics Centre, National Institute of Pathology - Indian Council of Medical Research, New Delhi, India
| |
Collapse
|
42
|
Abstract
Artificial intelligence (AI) has emerged as a major frontier in computer science research. Although AI has been available for some time and found its application in many fields of medicine, its use in dermatology is comparatively new and limited. A sound understanding of the concepts of AI is essential for dermatologists as skin conditions with their abundant clinical and dermatoscopic data and images can potentially be the next big thing in the application of AI in medicine. There are already a number of artificial intelligence studies focusing on skin disorders, such as skin cancer, psoriasis, atopic dermatitis and onychomycosis. This article presents an overview of AI and new developments relevant to dermatology, examining both its current applications and future potential.
Collapse
Affiliation(s)
- Abhishek De
- Department of Dermatology, Calcutta National Medical College, Kolkata, West Bengal, India
| | - Aarti Sarda
- Wizderm Specialty Skin and Hair Clinic, Kolkata, West Bengal, India
| | - Sachi Gupta
- Department of Dermatology, Hertford County Hospital, England
| | - Sudip Das
- Department of Dermatology, Calcutta National Medical College, Kolkata, West Bengal, India
| |
Collapse
|
43
|
Lee SH, Han P, Hales RK, Voong KR, Noro K, Sugiyama S, Haller JW, McNutt TR, Lee J. Multi-view radiomics and dosiomics analysis with machine learning for predicting acute-phase weight loss in lung cancer patients treated with radiotherapy. Phys Med Biol 2020; 65:195015. [PMID: 32235058 DOI: 10.1088/1361-6560/ab8531] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We propose a multi-view data analysis approach using radiomics and dosiomics (R&D) texture features for predicting acute-phase weight loss (WL) in lung cancer radiotherapy. Baseline weight of 388 patients who underwent intensity modulated radiation therapy (IMRT) was measured between one month prior to and one week after the start of IMRT. Weight change between one week and two months after the commencement of IMRT was analyzed, and dichotomized at 5% WL. Each patient had a planning CT and contours of gross tumor volume (GTV) and esophagus (ESO). A total of 355 features including clinical parameter (CP), GTV and ESO (GTV&ESO) dose-volume histogram (DVH), GTV radiomics, and GTV&ESO dosiomics features were extracted. R&D features were categorized as first- (L1), second- (L2), higher-order (L3) statistics, and three combined groups, L1 + L2, L2 + L3 and L1 + L2 + L3. Multi-view texture analysis was performed to identify optimal R&D input features. In the training set (194 earlier patients), feature selection was performed using Boruta algorithm followed by collinearity removal based on variance inflation factor. Machine-learning models were developed using Laplacian kernel support vector machine (lpSVM), deep neural network (DNN) and their averaged ensemble classifiers. Prediction performance was tested on an independent test set (194 more recent patients), and compared among seven different input conditions: CP-only, DVH-only, R&D-only, DVH + CP, R&D + CP, R&D + DVH and R&D + DVH + CP. Combined GTV L1 + L2 + L3 radiomics and GTV&ESO L3 dosiomics were identified as optimal input features, which achieved the best performance with an ensemble classifier (AUC = 0.710), having statistically significantly higher predictability compared with DVH and/or CP features (p < 0.05). When this performance was compared to that with full R&D-only features which reflect traditional single-view data, there was a statistically significant difference (p < 0.05). Using optimized multi-view R&D input features is beneficial for predicting early WL in lung cancer radiotherapy, leading to improved performance compared to using conventional DVH and/or CP features.
Collapse
Affiliation(s)
- Sang Ho Lee
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Liu Z, Mi M, Li X, Zheng X, Wu G, Zhang L. A lncRNA prognostic signature associated with immune infiltration and tumour mutation burden in breast cancer. J Cell Mol Med 2020; 24:12444-12456. [PMID: 32967061 PMCID: PMC7687003 DOI: 10.1111/jcmm.15762] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/13/2020] [Accepted: 08/05/2020] [Indexed: 02/06/2023] Open
Abstract
Current studies have shown that long non-coding RNAs (lncRNAs) may serve as prognostic biomarkers in multiple cancers. Therefore, we postulated that expression patterns of multiple lncRNAs combined into a single signature could improve clinicopathological risk stratification and prediction of overall survival rate for breast cancer patients. Two algorithms, Least Absolute Shrinkage and Selector Operation (LASSO) and Support Vector Machine-Recursive Feature Elimination (SVM-RFE), were used to select candidate lncRNAs. Univariate and multivariate Cox regression analyses were employed to construct a seven-lncRNA signature for breast cancer. Stratified analysis revealed that the signature was significantly associated with multiple clinicopathological risk factors. For clinical use, we developed a nomogram model to predict overall survival and odds of death for breast cancer patients. Single-sample gene set enrichment analysis (ssGSEA), CIBERSORT algorithm and ESTIMATE method were employed to assess the relative immune cell infiltrations of each sample. Differentially infiltration of immune cells and diverse tumour mutation burden (TMB) scores might give rise to the efficacy of lncRNA signature for predicting the overall survival of patients. Correlation analysis implied that LINC01215 was associated with multiple immune-related signalling pathways. A seven-lncRNA prognostic signature is a reliable tool to predict the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Zijian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mi Mi
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqian Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liling Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Wang Y, Chen L, Ju L, Xiao Y, Wang X. Tumor mutational burden related classifier is predictive of response to PD-L1 blockade in locally advanced and metastatic urothelial carcinoma. Int Immunopharmacol 2020; 87:106818. [PMID: 32738594 DOI: 10.1016/j.intimp.2020.106818] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Immunotherapy has made encouraging progress in the treatment of urothelial carcinoma, but only a small percentage of patients respond effectively to the immune checkpoint blockade (ICB). Our study aims to develop a classifier could effectively predict the response to ICB. METHODS Support vector machines recursive feature elimination (SVM-RFE) algorithm was used to feature selection, then compared nine common binary classification algorithms through machine learning, we selected the classifier with the highest prediction performance (LASSO logistics classifier). Ten-fold cross-validation was used to avoid the overfitting effect. RESULTS We developed a classifier on a urothelial carcinoma cohort treated with PD-L1 inhibitor Atzolizumab (IMvigor210 cohort, n = 272) and calculated a tumor mutational burden-related LASSO score (TLS) using the LASSO algorithm, which was significantly correlated with Tumor mutational burden (TMB) and neoantigen burden. We validated the efficacy of TLS in predicting prognosis and immunotherapy benefit in internal (IMvigor210) and external validation set (TCGA-BLCA, n = 406), respectively. CONCLUSIONS After in-depth analysis, we provide a new idea for stratified treatment of such patients, that is, patients with high TLS should use ICB and also may benefit from hormone therapy, while patients with low TLS respond poorly to ICB and maybe benefit from targeting TGFβ.
Collapse
Affiliation(s)
- Yejinpeng Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Liang Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lingao Ju
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China; Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China; Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China; Research Center of Wuhan for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China; Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China; Human Genetics Resource Preservation Center of Hubei Province, Wuhan, China; Human Genetics Resource Preservation Center of Wuhan University, Wuhan, China; Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Research Center of Wuhan for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China; Medical Research Institute, Wuhan University, Wuhan, China; Research Center of Wuhan for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
46
|
Hogarty DT, Hogarty JP, Hewitt AW. Smartphone use in ophthalmology: What is their place in clinical practice? Surv Ophthalmol 2020; 65:250-262. [DOI: 10.1016/j.survophthal.2019.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/29/2019] [Accepted: 09/09/2019] [Indexed: 01/02/2023]
|
47
|
Schachtschneider KM, Welge ME, Auvil LS, Chaki S, Rund LA, Madsen O, Elmore MR, Johnson RW, Groenen MA, Schook LB. Altered Hippocampal Epigenetic Regulation Underlying Reduced Cognitive Development in Response to Early Life Environmental Insults. Genes (Basel) 2020; 11:genes11020162. [PMID: 32033187 PMCID: PMC7074491 DOI: 10.3390/genes11020162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is involved in learning and memory and undergoes significant growth and maturation during the neonatal period. Environmental insults during this developmental timeframe can have lasting effects on brain structure and function. This study assessed hippocampal DNA methylation and gene transcription from two independent studies reporting reduced cognitive development stemming from early life environmental insults (iron deficiency and porcine reproductive and respiratory syndrome virus (PRRSv) infection) using porcine biomedical models. In total, 420 differentially expressed genes (DEGs) were identified between the reduced cognition and control groups, including genes involved in neurodevelopment and function. Gene ontology (GO) terms enriched for DEGs were associated with immune responses, angiogenesis, and cellular development. In addition, 116 differentially methylated regions (DMRs) were identified, which overlapped 125 genes. While no GO terms were enriched for genes overlapping DMRs, many of these genes are known to be involved in neurodevelopment and function, angiogenesis, and immunity. The observed altered methylation and expression of genes involved in neurological function suggest reduced cognition in response to early life environmental insults is due to altered cholinergic signaling and calcium regulation. Finally, two DMRs overlapped with two DEGs, VWF and LRRC32, which are associated with blood brain barrier permeability and regulatory T-cell activation, respectively. These results support the role of altered hippocampal DNA methylation and gene expression in early life environmentally-induced reductions in cognitive development across independent studies.
Collapse
Affiliation(s)
- Kyle M. Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60607, USA;
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Michael E. Welge
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Loretta S. Auvil
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
| | - Sulalita Chaki
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Laurie A. Rund
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Ole Madsen
- Animal Breeding and Genomics, Wageningen University, 6708 Wageningen, The Netherlands; (O.M.); (M.A.M.G.)
| | - Monica R.P. Elmore
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Rodney W. Johnson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
| | - Martien A.M. Groenen
- Animal Breeding and Genomics, Wageningen University, 6708 Wageningen, The Netherlands; (O.M.); (M.A.M.G.)
| | - Lawrence B. Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60607, USA;
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA; (M.E.W.); (L.S.A.)
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 616280, USA; (S.C.); (L.A.R.); (M.R.P.E.); (R.W.J.)
- Correspondence:
| |
Collapse
|
48
|
Hogarty DT, Su JC, Phan K, Attia M, Hossny M, Nahavandi S, Lenane P, Moloney FJ, Yazdabadi A. Artificial Intelligence in Dermatology-Where We Are and the Way to the Future: A Review. Am J Clin Dermatol 2020; 21:41-47. [PMID: 31278649 DOI: 10.1007/s40257-019-00462-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although artificial intelligence has been available for some time, it has garnered significant interest recently and has been popularized by major companies with its applications in image identification, speech recognition and problem solving. Artificial intelligence is now being increasingly studied for its potential uses in medicine. A sound understanding of the concepts of this emerging field is essential for the dermatologist as dermatology has abundant medical data and images that can be used to train artificial intelligence for patient care. There are already a number of artificial intelligence studies focusing on skin disorders such as skin cancer, psoriasis, atopic dermatitis and onychomycosis. This article aims to present a basic introduction to the concepts of artificial intelligence as well as present an overview of the current research into artificial intelligence in dermatology, examining both its current applications and its future potential.
Collapse
Affiliation(s)
- Daniel T Hogarty
- Monash University, Eastern Health, 5 Arnold Street, Box Hill 3128, Melbourne, VIC, Australia.
| | - John C Su
- Monash University, Eastern Health, 5 Arnold Street, Box Hill 3128, Melbourne, VIC, Australia
- Murdoch Children's Research Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin Phan
- Department of Dermatology, Liverpool Hospital, Sydney, NSW, Australia
| | - Mohamed Attia
- Institute for Intelligent Systems Research and Innovation, Geelong, Australia
| | - Mohammed Hossny
- Institute for Intelligent Systems Research and Innovation, Geelong, Australia
| | - Saeid Nahavandi
- Institute for Intelligent Systems Research and Innovation, Geelong, Australia
| | - Patricia Lenane
- Mater Misericordiae University Hospital, Dublin, Ireland
- University College Dublin, School of Medicine and Medical Science, Dublin, Ireland
| | - Fergal J Moloney
- Mater Misericordiae University Hospital, Dublin, Ireland
- University College Dublin, School of Medicine and Medical Science, Dublin, Ireland
| | - Anousha Yazdabadi
- Monash University, Eastern Health, 5 Arnold Street, Box Hill 3128, Melbourne, VIC, Australia
- Department of Dermatology, University of Melbourne, Melbourne, VIC, Australia
- Department of Dermatology, Northern Health, Epping, VIC, Australia
- Department of Dermatology, Deakin University, Melbourne, VIC, Australia
| |
Collapse
|
49
|
Naorem LD, Prakash VS, Muthaiyan M, Venkatesan A. Comprehensive analysis of dysregulated lncRNAs and their competing endogenous RNA network in triple-negative breast cancer. Int J Biol Macromol 2020; 145:429-436. [DOI: 10.1016/j.ijbiomac.2019.12.196] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/21/2019] [Accepted: 12/21/2019] [Indexed: 01/24/2023]
|
50
|
Review-Current Concepts in Inflammatory Skin Diseases Evolved by Transcriptome Analysis: In-Depth Analysis of Atopic Dermatitis and Psoriasis. Int J Mol Sci 2020; 21:ijms21030699. [PMID: 31973112 PMCID: PMC7037913 DOI: 10.3390/ijms21030699] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/14/2022] Open
Abstract
During the last decades, high-throughput assessment of gene expression in patient tissues using microarray technology or RNA-Seq took center stage in clinical research. Insights into the diversity and frequency of transcripts in healthy and diseased conditions provide valuable information on the cellular status in the respective tissues. Growing with the technique, the bioinformatic analysis toolkit reveals biologically relevant pathways which assist in understanding basic pathophysiological mechanisms. Conventional classification systems of inflammatory skin diseases rely on descriptive assessments by pathologists. In contrast to this, molecular profiling may uncover previously unknown disease classifying features. Thereby, treatments and prognostics of patients may be improved. Furthermore, disease models in basic research in comparison to the human disease can be directly validated. The aim of this article is not only to provide the reader with information on the opportunities of these techniques, but to outline potential pitfalls and technical limitations as well. Major published findings are briefly discussed to provide a broad overview on the current findings in transcriptomics in inflammatory skin diseases.
Collapse
|