1
|
Cao Z, Zhao S, Wu T, Sun F, Hu S, Shi L. Potential of gut microbiota metabolites in treating COPD: network pharmacology and Mendelian randomization approaches. Front Microbiol 2024; 15:1416651. [PMID: 39654679 PMCID: PMC11625750 DOI: 10.3389/fmicb.2024.1416651] [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: 04/12/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Objective The gut microbiota and its metabolites exert a significant influence on COPD, yet the underlying mechanisms remain elusive. We aim to holistically evaluate the role and mechanisms of the gut microbiota and its metabolites in COPD through network pharmacology and Mendelian randomization approaches. Methods Employing network pharmacology, we identified the gut microbiota and its metabolites' impact on COPD-related targets, elucidating the complex network mechanisms involving the gut microbiota, its metabolites, targets, and signaling pathways in relation to COPD. Further, promising gut microbiota metabolites and microbiota were pinpointed, with their causal relationships inferred through Mendelian randomization. Results A complex biological network was constructed, comprising 39 gut microbiota, 20 signaling pathways, 19 targets, and 23 metabolites associated with COPD. Phenylacetylglutamine emerged as a potentially promising metabolite for COPD treatment, with Mendelian randomization analysis revealing a causal relationship with COPD. Conclusion This study illuminates the intricate associations between the gut microbiota, its metabolites, and COPD. Phenylacetylglutamine may represent a novel avenue for COPD treatment. These findings could aid in identifying individuals at high risk for COPD, offering insights into early prevention and treatment strategies.
Collapse
Affiliation(s)
- Zhenghua Cao
- Graduate School, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Shengkun Zhao
- Graduate School, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Tong Wu
- Geriatric Department, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Feng Sun
- Respiratory Disease Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Shaodan Hu
- Respiratory Disease Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Li Shi
- Respiratory Disease Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
2
|
Nahali S, Safari L, Khanteymoori A, Huang J. StructmRNA a BERT based model with dual level and conditional masking for mRNA representation. Sci Rep 2024; 14:26043. [PMID: 39472486 PMCID: PMC11522565 DOI: 10.1038/s41598-024-77172-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 10/21/2024] [Indexed: 11/02/2024] Open
Abstract
In this study, we introduce StructmRNA, a new BERT-based model that was designed for the detailed analysis of mRNA sequences and structures. The success of DNABERT in understanding the intricate language of non-coding DNA with bidirectional encoder representations is extended to mRNA with StructmRNA. This new model uses a special dual-level masking technique that covers both sequence and structure, along with conditional masking. This enables StructmRNA to adeptly generate meaningful embeddings for mRNA sequences, even in the absence of explicit structural data, by capitalizing on the intricate sequence-structure correlations learned during extensive pre-training on vast datasets. Compared to well-known models like those in the Stanford OpenVaccine project, StructmRNA performs better in important tasks such as predicting RNA degradation. Thus, StructmRNA can inform better RNA-based treatments by predicting the secondary structures and biological functions of unseen mRNA sequences. The proficiency of this model is further confirmed by rigorous evaluations, revealing its unprecedented ability to generalize across various organisms and conditions, thereby marking a significant advance in the predictive analysis of mRNA for therapeutic design. With this work, we aim to set a new standard for mRNA analysis, contributing to the broader field of genomics and therapeutic development.
Collapse
Affiliation(s)
- Sepideh Nahali
- Information Retrieval and Knowledge Management Research Lab, York University, Toronto, Ontario, Canada.
- Department of Computer Engineering, University of Zanjan, Zanjan, Iran.
| | - Leila Safari
- Department of Computer Engineering, University of Zanjan, Zanjan, Iran
| | | | - Jimmy Huang
- Information Retrieval and Knowledge Management Research Lab, York University, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Li X, Heirman CC, Rickard AG, Sotolongo G, Castillo R, Adanlawo T, Everitt JI, Hodgin JB, Watts TL, Janowczyk A, Mowery YM, Barisoni L, Lafata KJ. Computational staining of CD3/CD20 positive lymphocytes in human tissues with experimental confirmation in a genetically engineered mouse model. Front Immunol 2024; 15:1451261. [PMID: 39530103 PMCID: PMC11550988 DOI: 10.3389/fimmu.2024.1451261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Immune dysregulation plays a major role in cancer progression. The quantification of lymphocytic spatial inflammation may enable spatial system biology, improve understanding of therapeutic resistance, and contribute to prognostic imaging biomarkers. Methods In this paper, we propose a knowledge-guided deep learning framework to measure the lymphocytic spatial architecture on human H&E tissue, where the fidelity of training labels is maximized through single-cell resolution image registration of H&E to IHC. We demonstrate that such an approach enables pixel-perfect ground-truth labeling of lymphocytes on H&E as measured by IHC. We then experimentally validate our technique in a genetically engineered, immune-compromised Rag2 mouse model, where Rag2 knockout mice lacking mature lymphocytes are used as a negative experimental control. Such experimental validation moves beyond the classical statistical testing of deep learning models and demonstrates feasibility of more rigorous validation strategies that integrate computational science and basic science. Results Using our developed approach, we automatically annotated more than 111,000 human nuclei (45,611 CD3/CD20 positive lymphocytes) on H&E images to develop our model, which achieved an AUC of 0.78 and 0.71 on internal hold-out testing data and external testing on an independent dataset, respectively. As a measure of the global spatial architecture of the lymphocytic microenvironment, the average structural similarity between predicted lymphocytic density maps and ground truth lymphocytic density maps was 0.86 ± 0.06 on testing data. On experimental mouse model validation, we measured a lymphocytic density of 96.5 ± %1% in a Rag2+/- control mouse, compared to an average of 16.2 ± %5% in Rag2-/- immune knockout mice (p<0.0001, ANOVA-test). Discussion These results demonstrate that CD3/CD20 positive lymphocytes can be accurately detected and characterized on H&E by deep learning and generalized across species. Collectively, these data suggest that our understanding of complex biological systems may benefit from computationally-derived spatial analysis, as well as integration of computational science and basic science.
Collapse
Affiliation(s)
- Xiang Li
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, United States
| | - Casey C. Heirman
- Medical Physics Graduate Program, Duke University, Durham, NC, United States
| | - Ashlyn G. Rickard
- Department of Radiation Oncology, Duke University, Durham, NC, United States
| | - Gina Sotolongo
- Department of Pathology, Duke University, Durham, NC, United States
| | - Rico Castillo
- Department of Radiation Oncology, Duke University, Durham, NC, United States
| | - Temitayo Adanlawo
- Department of Radiation Oncology, Duke University, Durham, NC, United States
| | | | - Jeffery B. Hodgin
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Tammara L. Watts
- Department of Head and Neck Surgery & Communication Sciences, Duke University, Durham, NC, United States
| | - Andrew Janowczyk
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, United States
- Department of Oncology, Division of Precision Oncology, Geneva University Hospitals, Geneva, Switzerland
- Department of Diagnostics, Division of Clinical Pathology, Geneva University Hospitals, Geneva, Switzerland
| | - Yvonne M. Mowery
- Department of Radiation Oncology, Duke University, Durham, NC, United States
- Department of Head and Neck Surgery & Communication Sciences, Duke University, Durham, NC, United States
- Department of Radiation Oncology, UPMC Hillman Cancer Center/University of Pittsburgh, Pittsburgh, PA, United States
| | - Laura Barisoni
- Department of Pathology, Duke University, Durham, NC, United States
- Department of Medicine, Division of Nephrology, Duke University, Durham, NC, United States
| | - Kyle J. Lafata
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, United States
- Medical Physics Graduate Program, Duke University, Durham, NC, United States
- Department of Radiation Oncology, Duke University, Durham, NC, United States
- Department of Pathology, Duke University, Durham, NC, United States
- Department of Radiology, Duke University, Durham, NC, United States
| |
Collapse
|
4
|
Zhai Y, Hai D, Zeng L, Lin C, Tan X, Mo Z, Tao Q, Li W, Xu X, Zhao Q, Shuai J, Pan J. Artificial intelligence-based evaluation of prognosis in cirrhosis. J Transl Med 2024; 22:933. [PMID: 39402630 PMCID: PMC11475999 DOI: 10.1186/s12967-024-05726-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Cirrhosis represents a significant global health challenge, characterized by high morbidity and mortality rates that severely impact human health. Timely and precise prognostic assessments of liver cirrhosis are crucial for improving patient outcomes and reducing mortality rates as they enable physicians to identify high-risk patients and implement early interventions. This paper features a thorough literature review on the prognostic assessment of liver cirrhosis, aiming to summarize and delineate the present status and constraints associated with the application of traditional prognostic tools in clinical settings. Among these tools, the Child-Pugh and Model for End-Stage Liver Disease (MELD) scoring systems are predominantly utilized. However, their accuracy varies significantly. These systems are generally suitable for broad assessments but lack condition-specific applicability and fail to capture the risks associated with dynamic changes in patient conditions. Future research in this field is poised for deep exploration into the integration of artificial intelligence (AI) with routine clinical and multi-omics data in patients with cirrhosis. The goal is to transition from static, unimodal assessment models to dynamic, multimodal frameworks. Such advancements will not only improve the precision of prognostic tools but also facilitate personalized medicine approaches, potentially revolutionizing clinical outcomes.
Collapse
Affiliation(s)
- Yinping Zhai
- Department of Gastroenterology Nursing Unit, Ward 192, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Darong Hai
- The School of Nursing, Wenzhou Medical University, Wenzhou, 325000, China
| | - Li Zeng
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325000, China
| | - Chenyan Lin
- The School of Nursing, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xinru Tan
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zefei Mo
- School of Biomedical Engineering, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Qijia Tao
- The School of Nursing, Wenzhou Medical University, Wenzhou, 325000, China
| | - Wenhui Li
- The School of Nursing, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaowei Xu
- Department of Gastroenterology Nursing Unit, Ward 192, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qi Zhao
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan, 114051, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China.
| | - Jianwei Shuai
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, 325000, China.
| | - Jingye Pan
- Department of Big Data in Health Science, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, 325000, China.
- Zhejiang Engineering Research Center for Hospital Emergency and Process Digitization, Wenzhou, 325000, China.
| |
Collapse
|
5
|
Chen G, Jin Y, Chu C, Zheng Y, Chen Y, Zhu X. Genetic prediction of blood metabolites mediating the relationship between gut microbiota and Alzheimer's disease: a Mendelian randomization study. Front Microbiol 2024; 15:1414977. [PMID: 39224217 PMCID: PMC11366617 DOI: 10.3389/fmicb.2024.1414977] [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: 04/10/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Background Observational studies have suggested an association between gut microbiota and Alzheimer's disease (AD); however, the causal relationship remains unclear, and the role of blood metabolites in this association remains elusive. Purpose To elucidate the causal relationship between gut microbiota and AD and to investigate whether blood metabolites serve as potential mediators. Materials and methods Univariable Mendelian randomization (UVMR) analysis was employed to assess the causal relationship between gut microbiota and AD, while multivariable MR (MVMR) was utilized to mitigate confounding factors. Subsequently, a two-step mediation MR approach was employed to explore the role of blood metabolites as potential mediators. We primarily utilized the inverse variance-weighted method to evaluate the causal relationship between exposure and outcome, and sensitivity analyses including Contamination mixture, Maximum-likelihood, Debiased inverse-variance weighted, MR-Egger, Bayesian Weighted Mendelian randomization, and MR pleiotropy residual sum and outlier were conducted to address pleiotropy. Results After adjustment for reverse causality and MVMR correction, class Actinobacteria (OR: 1.03, 95% CI: 1.01-1.06, p = 0.006), family Lactobacillaceae (OR: 1.03, 95% CI: 1.00-1.05, p = 0.017), genus Lachnoclostridium (OR: 1.03, 95% CI: 1.00-1.06, p = 0.019), genus Ruminiclostridium9 (OR: 0.97, 95% CI: 0.94-1.00, p = 0.027) and genus Ruminiclostridium6 (OR: 1.03, 95% CI: 1.01-1.05, p = 0.009) exhibited causal effects on AD. Moreover, 1-ribosyl-imidazoleacetate levels (-6.62%), Metabolonic lactone sulfate levels (2.90%), and Nonadecanoate (19:0) levels (-12.17%) mediated the total genetic predictive effects of class Actinobacteria on AD risk. Similarly, 2-stearoyl-GPE (18:0) levels (-9.87%), Octadecanedioylcarnitine (C18-DC) levels (4.44%), 1-(1-enyl-stearoyl)-2-oleoyl-GPE (p-18:0/18:1) levels (38.66%), and X-23639 levels (13.28%) respectively mediated the total genetic predictive effects of family Lactobacillaceae on AD risk. Furthermore, Hexadecanedioate (C16-DC) levels (5.45%) mediated the total genetic predictive effects of genus Ruminiclostridium 6 on AD risk; Indole-3-carboxylate levels (13.91%), X-13431 levels (7.08%), Alpha-ketoglutarate to succinate ratio (-13.91%), 3-phosphoglycerate to glycerate ratio (15.27%), and Succinate to proline ratio (-14.64%) respectively mediated the total genetic predictive effects of genus Ruminiclostridium 9 on AD risk. Conclusion Our mediation MR analysis provides genetic evidence suggesting the potential mediating role of blood metabolites in the causal relationship between gut microbiota and AD. Further large-scale randomized controlled trials are warranted to validate the role of blood metabolites in the specific mechanisms by which gut microbiota influence AD.
Collapse
Affiliation(s)
- Guanglei Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yaxian Jin
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Cancan Chu
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yuhao Zheng
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yunzhi Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xing Zhu
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
6
|
Shen Y, Li C, Zhang X, Wang Y, Zhang H, Yu Z, Gui B, Hu R, Li Q, Gao A, Liang H. Gut microbiota linked to hydrocephalus through inflammatory factors: a Mendelian randomization study. Front Immunol 2024; 15:1372051. [PMID: 39076985 PMCID: PMC11284128 DOI: 10.3389/fimmu.2024.1372051] [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: 01/17/2024] [Accepted: 06/27/2024] [Indexed: 07/31/2024] Open
Abstract
Background The gut microbiota (GM) has been implicated in neurological disorders, but the relationship with hydrocephalus, especially the underlying mechanistic pathways, is unclear. Using Mendelian randomization (MR), we aim to discover the mediating role of inflammatory factors in the relationship between GM and hydrocephalus. Methods After removing confounders, univariable and multivariable MR analyses were performed using summary statistics to assess the causal relationships between GM, inflammatory factors (IL-17A and IL-27), and types of hydrocephalus. Meta-analyses were used to reconcile the differences in MR results between different hydrocephalus sources. Finally, mediator MR analyses were applied to determine the mediating effect of inflammatory factors. Various sensitivity analysis methods were employed to ensure the reliability and stability of the results. Results After correction for P-values, Firmicutes (phylum) (OR, 0.34; 95%CI, 0.17-0.69; P = 2.71E-03, P FDR = 2.44E-02) significantly reduced the risk of obstructive hydrocephalus. The remaining 18 different taxa of GM had potential causal relationships for different types of hydrocephalus. In addition, Firmicutes (phylum) decreased the risk of obstructive hydrocephalus by increasing levels of IL-17A (mediating effect = 21.01%), while Eubacterium ruminantium group (genus) increased the risk of normal-pressure hydrocephalus by decreasing levels of IL-27 (mediating effect = 7.48%). Conclusion We reveal the connection between GM, inflammatory factors (IL-17A and IL-27), and hydrocephalus, which lays the foundation for unraveling the mechanism between GM and hydrocephalus.
Collapse
Affiliation(s)
- Yingjie Shen
- Department of Neurosurgery, National Health Commission Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Changyu Li
- Department of Neurosurgery, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Xi Zhang
- Department of Neurosurgery, National Health Commission Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yaolou Wang
- Department of Neurosurgery, National Health Commission Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Haopeng Zhang
- Department of Neurosurgery, National Health Commission Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhao Yu
- Department of Neurosurgery, National Health Commission Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Binbin Gui
- Department of Neurosurgery, National Health Commission Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Renjie Hu
- Department of Neurosurgery, National Health Commission Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qi Li
- Department of Neurosurgery, National Health Commission Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Aili Gao
- School of Life Science, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Hongsheng Liang
- Department of Neurosurgery, National Health Commission Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
7
|
Liu C, Wu H, Li K, Chi Y, Wu Z, Xing C. Identification of biomarkers for abdominal aortic aneurysm in Behçet's disease via mendelian randomization and integrated bioinformatics analyses. J Cell Mol Med 2024; 28:e18398. [PMID: 38785203 PMCID: PMC11117452 DOI: 10.1111/jcmm.18398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/03/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
Behçet's disease (BD) is a complex autoimmune disorder impacting several organ systems. Although the involvement of abdominal aortic aneurysm (AAA) in BD is rare, it can be associated with severe consequences. In the present study, we identified diagnostic biomarkers in patients with BD having AAA. Mendelian randomization (MR) analysis was initially used to explore the potential causal association between BD and AAA. The Limma package, WGCNA, PPI and machine learning algorithms were employed to identify potential diagnostic genes. A receiver operating characteristic curve (ROC) for the nomogram was constructed to ascertain the diagnostic value of AAA in patients with BD. Finally, immune cell infiltration analyses and single-sample gene set enrichment analysis (ssGSEA) were conducted. The MR analysis indicated a suggestive association between BD and the risk of AAA (odds ratio [OR]: 1.0384, 95% confidence interval [CI]: 1.0081-1.0696, p = 0.0126). Three hub genes (CD247, CD2 and CCR7) were identified using the integrated bioinformatics analyses, which were subsequently utilised to construct a nomogram (area under the curve [AUC]: 0.982, 95% CI: 0.944-1.000). Finally, the immune cell infiltration assay revealed that dysregulation immune cells were positively correlated with the three hub genes. Our MR analyses revealed a higher susceptibility of patients with BD to AAA. We used a systematic approach to identify three potential hub genes (CD247, CD2 and CCR7) and developed a nomogram to assist in the diagnosis of AAA among patients with BD. In addition, immune cell infiltration analysis indicated the dysregulation in immune cell proportions.
Collapse
Affiliation(s)
- Chunjiang Liu
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Huadong Wu
- Department of vascular surgeryFirst affiliated Hospital of Huzhou UniversityHuzhouChina
| | - Kuan Li
- Department of General SurgeryKunshan Hospital of Traditional Chinese MedicineKunshanChina
| | - Yongxing Chi
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Zhaoying Wu
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Chungen Xing
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
8
|
Fu H, Xu T, Zhao W, Jiang L, Shan S. Roles of gut microbiota in androgenetic alopecia: insights from Mendelian randomization analysis. Front Microbiol 2024; 15:1360445. [PMID: 38628866 PMCID: PMC11018880 DOI: 10.3389/fmicb.2024.1360445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Background Androgenetic alopecia (AGA) is the most common type of androgen-associated hair loss. Previous studies have indicated an association between the gut microbiota and AGA. To delve deeper, we executed a two-sample Mendelian randomization (MR) analysis to investigate the potential causal relationship between the gut microbiota and AGA. Methods A two-sample MR investigation was utilized to delve into the intricate interplay between gut microbiota and AGA. Information regarding 211 gut microbial taxa was sourced from the MiBioGen consortium. The summary statistics of the genome-wide association studies (GWAS) for AGA were obtained from the FinnGen biobank, which included 195 cases and 201,019 controls. Various analytical approaches, including Inverse Variance Weighting (IVW), Weighted Median, MR-Egger, Weighted Mode, and Simple Mode were employed to evaluate the causal impact of gut microbiota on AGA. Sensitivity analyses were subsequently conducted to affirm the robustness of the findings. Results A two-sample MR investigation unveiled the genus Olsenella, genus Ruminococcaceae UCG-004, and genus Ruminococcaceae UCG-010 were identified as risk factors associated with AGA. In contrast, the family Acidaminococcaceae and genus Anaerofilum, along with the genus Ruminiclostridium 9, demonstrated a protective effect. The sensitivity analyses provided additional assurance that the findings of the current study were less susceptible to the influence of confounding variables and biases. Conclusion The MR study has established a link between specific gut microbiota and AGA, offering evidence for the identification of more precisely targeted probiotics. This discovery has the potential to aid in the prevention, control, and reversal of AGA progression.
Collapse
Affiliation(s)
- Haijing Fu
- Department of Dermatology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Tianyi Xu
- Department of Dermatology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wumei Zhao
- Department of Dermatology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Leiwei Jiang
- Department of Dermatology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Shijun Shan
- Department of Dermatology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Hangzhou Third People’s Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Mortezaee K. WNT/β-catenin regulatory roles on PD-(L)1 and immunotherapy responses. Clin Exp Med 2024; 24:15. [PMID: 38280119 PMCID: PMC10822012 DOI: 10.1007/s10238-023-01274-z] [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: 10/18/2023] [Accepted: 11/29/2023] [Indexed: 01/29/2024]
Abstract
Dysregulation of WNT/β-catenin is a hallmark of many cancer types and a key mediator of metastasis in solid tumors. Overactive β-catenin signaling hampers dendritic cell (DC) recruitment, promotes CD8+ T cell exclusion and increases the population of regulatory T cells (Tregs). The activity of WNT/β-catenin also induces the expression of programmed death-ligand 1 (PD-L1) on tumor cells and promotes programmed death-1 (PD-1) upregulation. Increased activity of WNT/β-catenin signaling after anti-PD-1 therapy is indicative of a possible implication of this signaling in bypassing immune checkpoint inhibitor (ICI) therapy. This review is aimed at giving a comprehensive overview of the WNT/β-catenin regulatory roles on PD-1/PD-L1 axis in tumor immune ecosystem, discussing about key mechanistic events contributed to the WNT/β-catenin-mediated bypass of ICI therapy, and representing inhibitors of this signaling as promising combinatory regimen to go with anti-PD-(L)1 in cancer immunotherapy. Ideas presented in this review imply the synergistic efficacy of such combination therapy in rendering durable anti-tumor immunity.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
10
|
Nørgård MØ, Svenningsen P. Acute Kidney Injury by Ischemia/Reperfusion and Extracellular Vesicles. Int J Mol Sci 2023; 24:15312. [PMID: 37894994 PMCID: PMC10607034 DOI: 10.3390/ijms242015312] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Acute kidney injury (AKI) is often caused by ischemia-reperfusion injury (IRI). IRI significantly affects kidney metabolism, which elicits pro-inflammatory responses and kidney injury. The ischemia/reperfusion of the kidney is associated with transient high mitochondrial-derived reactive oxygen species (ROS) production rates. Excessive mitochondrial-derived ROS damages cellular components and, together with other pathogenic mechanisms, elicits a range of acute injury mechanisms that impair kidney function. Mitochondrial-derived ROS production also stimulates epithelial cell secretion of extracellular vesicles (EVs) containing RNAs, lipids, and proteins, suggesting that EVs are involved in AKI pathogenesis. This literature review focuses on how EV secretion is stimulated during ischemia/reperfusion and how cell-specific EVs and their molecular cargo may modify the IRI process. Moreover, critical pitfalls in the analysis of kidney epithelial-derived EVs are described. In particular, we will focus on how the release of kidney epithelial EVs is affected during tissue analyses and how this may confound data on cell-to-cell signaling. By increasing awareness of methodological pitfalls in renal EV research, the risk of false negatives can be mitigated. This will improve future EV data interpretation regarding EVs contribution to AKI pathogenesis and their potential as biomarkers or treatments for AKI.
Collapse
Affiliation(s)
| | - Per Svenningsen
- Department of Molecular Medicine, University of Southern Denmark, DK-5000 Odense, Denmark;
| |
Collapse
|
11
|
Plasma Extracellular Vesicle Long RNA in Diagnosis and Prediction in Small Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14225493. [PMID: 36428585 PMCID: PMC9688902 DOI: 10.3390/cancers14225493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/23/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
(1) Introduction: The aim of this study was to identify the plasma extracellular vesicle (EV)-specific transcriptional profile in small-cell lung cancer (SCLC) and to explore the application value of plasma EV long RNA (exLR) in SCLC treatment prediction and diagnosis. (2) Methods: Plasma samples were collected from 57 SCLC treatment-naive patients, 104 non-small-cell lung cancer (NSCLC) patients and 59 healthy participants. The SCLC patients were divided into chemo-sensitive and chemo-refractory groups based on the therapeutic effects. The exLR profiles of the plasma samples were analyzed by high-throughput sequencing. Bioinformatics approaches were used to investigate the differentially expressed exLRs and their biofunctions. Finally, a t-signature was constructed using logistic regression for SCLC treatment prediction and diagnosis. (3) Results: We obtained 220 plasma exLRs profiles in all the participants. Totals of 5787 and 1207 differentially expressed exLRs were identified between SCLC/healthy controls, between the chemo-sensitive/chemo-refractory groups, respectively. Furthermore, we constructed a t-signature that comprised ten exLRs, including EPCAM, CCNE2, CDC6, KRT8, LAMB1, CALB2, STMN1, UCHL1, HOXB7 and CDCA7, for SCLC treatment prediction and diagnosis. The exLR t-score effectively distinguished the chemo-sensitive from the chemo-refractory group (p = 9.268 × 10-9) with an area under the receiver operating characteristic curve (AUC) of 0.9091 (95% CI: 0.837 to 0.9811) and distinguished SCLC from healthy controls (AUC: 0.9643; 95% CI: 0.9256-1) and NSCLC (AUC: 0.721; 95% CI: 0.6384-0.8036). (4) Conclusions: This study firstly characterized the plasma exLR profiles of SCLC patients and verified the feasibility and value of identifying biomarkers based on exLR profiles in SCLC diagnosis and treatment prediction.
Collapse
|
12
|
Auber M, Svenningsen P. An estimate of extracellular vesicle secretion rates of human blood cells. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e46. [PMID: 38938292 PMCID: PMC11080926 DOI: 10.1002/jex2.46] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/26/2022] [Accepted: 05/11/2022] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) have been implicated in the intercellular transfer of RNA and proteins through cellular secretion into the extracellular space. In blood plasma, circulating EVs are mainly derived from blood cells; however, factors that control plasma EV abundance are largely unknown. Here, we estimate the EV secretion rates for blood cell types using reported values for cell-specific plasma EV abundances and their parental cell's ubiquity in healthy humans. While we found that plasma contains on average ∼2 plasma EVs/cell, the cell-specific EV-to-cell ratio spanned four orders of magnitude from 0.13 ± 0.1 erythrocyte-derived EVs/erythrocyte to (1.9 ± 1.3) × 103 monocyte-derived EVs/monocyte. The steady-state plasma EV level was maintained by an estimated plasma EV secretion rate of (1.5 ± 0.4) × 1012 EVs/min. The cell-specific secretion rate estimates were highest for monocytes (45 ± 21 EVs/cell/min) and lowest for erythrocytes ((3.2 ± 3.0) × 10-3 EVs/cell/min). The estimated basal cell-specific EV secretion rates were not significantly correlated to the cell's lifespan or size; however, we observed a highly significant correlation to cellular mitochondrial enzyme activities. Together, our analysis indicates that cell-specific mitochondrial metabolism, for example, via reactive oxygen species, affects plasma EV abundance through increased secretion rates, and the results provide a resource for understanding EV function in human health and disease.
Collapse
Affiliation(s)
- Martin Auber
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Per Svenningsen
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| |
Collapse
|
13
|
Du C, Liu X, Li M, Zhao Y, Li J, Wen Z, Liu M, Yang M, Fu B, Wei M. Analysis of 5-Methylcytosine Regulators and DNA Methylation-Driven Genes in Colon Cancer. Front Cell Dev Biol 2022; 9:657092. [PMID: 35174154 PMCID: PMC8842075 DOI: 10.3389/fcell.2021.657092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 12/17/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Epigenetic-driven events are important molecular mechanisms of carcinogenesis. The 5-methylcytosine (5mC) regulators play important roles in the methylation-driven gene expression. However, the effect of the 5mC regulators on the oncogenic pathways in colon cancer (CC) remains unclear. Also, the clinical value of such epigenetic-driven events needs further research. Methods: The transcriptome and matching epigenetic data were obtained from The Cancer Genome Atlas dataset. The gene set variation analysis identified the oncogenic pathways adjusted by 5mC regulators. The “edgeR” and “methylmix” package identified the differential expression genes of DNA methylation-driven genes. The correlation between 5mC regulators or transcription factors and shortlisted genes was investigated by calculating the Spearman's rank correlation coefficient. Among them, the genes related to diagnosis were screened out based on differential gene expression in extracellular vesicles (EVs) by the “limma” package and histology by immunohistochemistry. Then, a risk signature was constructed by fitting the generalized linear model and validated by the receiver operating characteristic curve. Results: MYC targets pathway and phosphatidylinositol-3-kinase–AKT–mammalian target of rapamycin signaling pathway were identified as the hallmark-related pathways associated with 5mC regulators. Also, the P53 pathway was subject to the influence of regulators' expression. A five methylation-driven gene signature (FIRRE, MYBL2, TGFBI, AXIN2, and SLC35D3) was developed as the biomarker for CC diagnosis. Meanwhile, those genes positively related to 5mC regulators and interacted with their relevant or transcription factors. Conclusion: In general, 5mC regulators are positively related to each other and DNA methylation-driven genes, with the relationship of multiple active and inhibitory pathways related to cancer. Meanwhile, the signature (FIRRE, MYBL2, TGFBI, AXIN2, and SLC35D3) can prefigure prospective diagnosis in CC.
Collapse
Affiliation(s)
- Cheng Du
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - XinLi Liu
- Department of Digestive Oncology, Cancer Hospital of China Medical University, Shenyang, China
| | - Mingwei Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Yi Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Jie Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Zhikang Wen
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Min Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Meina Yang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Boshi Fu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Shenyang, China.,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|