1
|
Zhang X, Zhou CG, Ma LJ. Role of GDF-15 in diabetic nephropathy: mechanisms, diagnosis, and therapeutic potential. Int Urol Nephrol 2025; 57:169-175. [PMID: 39150600 DOI: 10.1007/s11255-024-04179-2] [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: 04/18/2024] [Accepted: 07/25/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Growth differentiation factor 15 (GDF-15) is a cytokine involved in regulating homeostasis, and its expression is up-regulated in response to injury, stress, and inflammation. This study explored the role of GDF-15 in diabetic nephropathy (DN), a severe complication of diabetes mellitus, and its potential as a biomarker for disease progression. METHODS As a member of the transforming growth factor-β superfamily, GDF-15 exhibits its renal protective functions primarily through its anti-inflammatory effects and the up-regulation of other renal protective factors. This study evaluated the association between circulating GDF-15 levels and DN progression, examining the underlying mechanisms. RESULTS Circulating GDF-15 levels are closely linked to the development and progression of DN. While existing research has yielded some consistent conclusions, a comprehensive understanding of the role of GDF-15 in DN pathogenesis is needed to identify new therapeutic targets and strategies. CONCLUSION GDF-15 has the potential to be a prognostic and diagnostic biomarker for DN. It is crucial to establish appropriate reference ranges and explore their clinical utility in routine practice for validating the role of GDF-15 in DN management. Further interventional studies are required to confirm its clinical value in diagnosing and predicting the progression of DN.
Collapse
Affiliation(s)
- Xin Zhang
- Clinical Laboratory, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201100, China
| | - Chen-Gang Zhou
- Clinical Laboratory, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201100, China
| | - Li-Jia Ma
- Clinical Laboratory, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201100, China.
- Pathology Department, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201100, China.
| |
Collapse
|
2
|
Delrue C, Speeckaert MM. Decoding Kidney Pathophysiology: Omics-Driven Approaches in Precision Medicine. J Pers Med 2024; 14:1157. [PMID: 39728069 DOI: 10.3390/jpm14121157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Chronic kidney disease (CKD) is a major worldwide health concern because of its progressive nature and complex biology. Traditional diagnostic and therapeutic approaches usually fail to account for disease heterogeneity, resulting in low efficacy. Precision medicine offers a novel approach to studying kidney disease by combining omics technologies such as genomics, transcriptomics, proteomics, metabolomics, and epigenomics. By identifying discrete disease subtypes, molecular biomarkers, and therapeutic targets, these technologies pave the way for personalized treatment approaches. Multi-omics integration has enhanced our understanding of CKD by revealing intricate molecular linkages and pathways that contribute to treatment resistance and disease progression. While pharmacogenomics offers insights into expected responses to personalized treatments, single-cell and spatial transcriptomics can be utilized to investigate biological heterogeneity. Despite significant development, challenges persist, including data integration concerns, high costs, and ethical quandaries. Standardized data protocols, collaborative data-sharing frameworks, and advanced computational tools such as machine learning and causal inference models are required to address these challenges. With the advancement of omics technology, nephrology may benefit from improved diagnostic accuracy, risk assessment, and personalized care. By overcoming these barriers, precision medicine has the potential to develop novel techniques for improving patient outcomes in kidney disease treatment.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, 9000 Ghent, Belgium
- Research Foundation-Flanders (FWO), 1000 Brussels, Belgium
| |
Collapse
|
3
|
Zhang Y, Yang YS, Chen WC, Wang CM, He HF. Constructing and Validating a Network of Potential Olfactory Sheathing Cell Transplants Regulating Spinal Cord Injury Progression. Mol Neurobiol 2023; 60:6883-6895. [PMID: 37515671 DOI: 10.1007/s12035-023-03510-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/10/2023] [Indexed: 07/31/2023]
Abstract
The pathology of spinal cord injury (SCI), including primary and secondary injuries, primarily involves hemorrhage, ischemia, edema, and inflammatory responses. Cell transplantation has been the most promising treatment for SCI in recent years; however, its specific molecular mechanism remains unclear. In this study, bioinformatics analysis verified by experiment was used to elucidate the hub genes associated with SCI and to discover the underlying molecular mechanisms of cell intervention. GSE46988 data were downloaded from the Gene Expression Omnibus dataset. In our study, differentially expressed genes (DEGs) were reanalyzed using the "R" software (R v4.2.1). Functional enrichment and protein-protein interaction network analyses were performed, and key modules and hub genes were identified. Network construction was performed for the hub genes and their associated miRNAs. Finally, a semi-quantitative analysis of hub genes and pathways was performed using quantitative real-time polymerase chain reaction. In total, 718 DEGs were identified, mainly enriched in immune and inflammation-related functions. We found that Cd4, Tp53, Rac2, and Akt3 differed between vehicle and transplanted groups, suggesting that these genes may play an essential role in the transplantation of olfactory ensheathing cells, while a toll-like receptor signaling pathway was significantly enriched in Gene set enrichment analysis, and then, the differences were statistically significant by experimentally verifying the expression of their associated molecules (Tlr4, Nf-κb, Ikkβ, Cxcl2, and Tnf-α). In addition, we searched for upstream regulatory molecules of these four central genes and constructed a regulatory network. This study is the first to construct a regulatory network for olfactory ensheathing cell transplantation in treating SCI, providing a new idea for SCI cell therapy.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yu-Shen Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Wei-Can Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Cong-Mei Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - He-Fan He
- Department of Anesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| |
Collapse
|
4
|
Li Z, Zeng N, Zhao X, Chen X, Liang G, Liu H, Lin J, Zheng P, Lin X, Zhou H, Zheng D. Urinary peptidome analysis in CKD and IgA nephropathy. Clin Kidney J 2023; 16:1965-1973. [PMID: 37915931 PMCID: PMC10616486 DOI: 10.1093/ckj/sfad211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Indexed: 11/03/2023] Open
Abstract
Background Chronic kidney disease (CKD) has emerged as a significant challenge to human health and economic stability in aging societies worldwide. Current clinical practice strategies remain insufficient for the early identification of kidney dysfunction, and the differential diagnosis of immunoglobulin A nephropathy (IgAN) predominantly relies on invasive kidney biopsy procedures. Methods First, we assessed a case-control cohort to obtain urine samples from healthy controls and biopsy-confirmed CKD patients. Matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometry (MS) was applied to detect urinary peptide and then these urinary peptide profiles were used to construct diagnostic models to distinguish CKD patients from controls and identify IgAN patients from other nephropathy patients. Furthermore, we assessed the robustness of the diagnostic models and their reproducibility by applying different algorithms. Results A rapid and accurate working platform for detecting CKD and its IgAN subtype based on urinary peptide pattern detected by MALDI-TOF MS was established. Naturally occurring urinary peptide profiles were used to construct a diagnostic model to distinguish CKD patients from controls and identify IgAN patients from other nephropathy patients. The performance of several algorithms was assessed and demonstrated that the robustness of the diagnostic models as well as their reproducibility were satisfactory. Conclusions The present findings suggest that the CKD-related and IgAN-specific urinary peptides discovered facilitate precise identification of CKD and its IgAN subtype, offering a dependable framework for screening conditions linked to renal dysfunction. This will aid in comprehending the pathogenesis of nephropathy and identifying potential protein targets for the clinical management of nephropathy.
Collapse
Affiliation(s)
- Zewen Li
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Nianyi Zeng
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Zhao
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xuedong Chen
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guangqing Liang
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haiyue Liu
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Division of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jinyan Lin
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peizhuang Zheng
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xingtao Lin
- Department of Pathology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Hongwei Zhou
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Daowen Zheng
- Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Gerontology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Kiernan E, Surapaneni A, Zhou L, Schlosser P, Walker KA, Rhee EP, Ballantyne CM, Deo R, Dubin RF, Ganz P, Coresh J, Grams ME. Alterations in the Circulating Proteome Associated with Albuminuria. J Am Soc Nephrol 2023; 34:1078-1089. [PMID: 36890639 PMCID: PMC10278823 DOI: 10.1681/asn.0000000000000108] [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: 09/08/2022] [Accepted: 02/05/2023] [Indexed: 03/10/2023] Open
Abstract
SIGNIFICANCE STATEMENT We describe circulating proteins associated with albuminuria in a population of African American Study of Kidney Disease and Hypertension with CKD (AASK) using the largest proteomic platform to date: nearly 7000 circulating proteins, representing approximately 2000 new targets. Findings were replicated in a subset of a general population cohort with kidney disease (ARIC) and a population with CKD Chronic Renal Insufficiency Cohort (CRIC). In cross-sectional analysis, 104 proteins were significantly associated with albuminuria in the Black group, of which 67 of 77 available proteins were replicated in ARIC and 68 of 71 available proteins in CRIC. LMAN2, TNFSFR1B, and members of the ephrin superfamily had the strongest associations. Pathway analysis also demonstrated enrichment of ephrin family proteins. BACKGROUND Proteomic techniques have facilitated understanding of pathways that mediate decline in GFR. Albuminuria is a key component of CKD diagnosis, staging, and prognosis but has been less studied than GFR. We sought to investigate circulating proteins associated with higher albuminuria. METHODS We evaluated the cross-sectional associations of the blood proteome with albuminuria and longitudinally with doubling of albuminuria in the African American Study of Kidney Disease and Hypertension (AASK; 38% female; mean GFR 46; median urine protein-to-creatinine ratio 81 mg/g; n =703) and replicated in two external cohorts: a subset of the Atherosclerosis Risk in Communities (ARIC) study with CKD and the Chronic Renal Insufficiency Cohort (CRIC). RESULTS In cross-sectional analysis, 104 proteins were significantly associated with albuminuria in AASK, of which 67 of 77 available proteins were replicated in ARIC and 68 of 71 available proteins in CRIC. Proteins with the strongest associations included LMAN2, TNFSFR1B, and members of the ephrin superfamily. Pathway analysis also demonstrated enrichment of ephrin family proteins. Five proteins were significantly associated with worsening albuminuria in AASK, including LMAN2 and EFNA4, which were replicated in ARIC and CRIC. CONCLUSIONS Among individuals with CKD, large-scale proteomic analysis identified known and novel proteins associated with albuminuria and suggested a role for ephrin signaling in albuminuria progression.
Collapse
Affiliation(s)
- Elizabeth Kiernan
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aditya Surapaneni
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, New York
| | - Linda Zhou
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Pascal Schlosser
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, Baltimore, Maryland
| | - Eugene P. Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ruth F. Dubin
- Division of Nephrology, University of Texas—Southwestern, Dallas, Texas
| | - Peter Ganz
- Division of Cardiology, Zuckerberg San Francisco General Hospital and Department of Medicine, University of California San Francisco, San Francisco, California
| | - Josef Coresh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Morgan E. Grams
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
- Division of Precision Medicine, New York University Grossman School of Medicine, New York, New York
| |
Collapse
|
6
|
Sirolli V, Piscitani L, Bonomini M. Biomarker-Development Proteomics in Kidney Transplantation: An Updated Review. Int J Mol Sci 2023; 24:ijms24065287. [PMID: 36982359 PMCID: PMC10049725 DOI: 10.3390/ijms24065287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Kidney transplantation (KT) is the optimal therapeutic strategy for patients with end-stage renal disease. The key to post-transplantation management is careful surveillance of allograft function. Kidney injury may occur from several different causes that require different patient management approaches. However, routine clinical monitoring has several limitations and detects alterations only at a later stage of graft damage. Accurate new noninvasive biomarker molecules are clearly needed for continuous monitoring after KT in the hope that early diagnosis of allograft dysfunction will lead to an improvement in the clinical outcome. The advent of “omics sciences”, and in particular of proteomic technologies, has revolutionized medical research. Proteomic technologies allow us to achieve the identification, quantification, and functional characterization of proteins/peptides in biological samples such as urine or blood through supervised or targeted analysis. Many studies have investigated proteomic techniques as potential molecular markers discriminating among or predicting allograft outcomes. Proteomic studies in KT have explored the whole transplant process: donor, organ procurement, preservation, and posttransplant surgery. The current article reviews the most recent findings on proteomic studies in the setting of renal transplantation in order to better understand the effective potential of this new diagnostic approach.
Collapse
Affiliation(s)
- Vittorio Sirolli
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS. Annunziata Hospital, 66013 Chieti, Italy
| | - Luca Piscitani
- Nephrology and Dialysis Unit, Department of Medicine, San Salvatore Hospital, 67100 L’Aquila, Italy
| | - Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS. Annunziata Hospital, 66013 Chieti, Italy
- Correspondence:
| |
Collapse
|
7
|
He B, Huang Z, Huang C, Nice EC. Clinical applications of plasma proteomics and peptidomics: Towards precision medicine. Proteomics Clin Appl 2022; 16:e2100097. [PMID: 35490333 DOI: 10.1002/prca.202100097] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/16/2022] [Accepted: 04/28/2022] [Indexed: 02/05/2023]
Abstract
In the context of precision medicine, disease treatment requires individualized strategies based on the underlying molecular characteristics to overcome therapeutic challenges posed by heterogeneity. For this purpose, it is essential to develop new biomarkers to diagnose, stratify, or possibly prevent diseases. Plasma is an available source of biomarkers that greatly reflects the physiological and pathological conditions of the body. An increasing number of studies are focusing on proteins and peptides, including many involving the Human Proteome Project (HPP) of the Human Proteome Organization (HUPO), and proteomics and peptidomics techniques are emerging as critical tools for developing novel precision medicine preventative measures. Excitingly, the emerging plasma proteomics and peptidomics toolbox exhibits a huge potential for studying pathogenesis of diseases (e.g., COVID-19 and cancer), identifying valuable biomarkers and improving clinical management. However, the enormous complexity and wide dynamic range of plasma proteins makes plasma proteome profiling challenging. Herein, we summarize the recent advances in plasma proteomics and peptidomics with a focus on their emerging roles in COVID-19 and cancer research, aiming to emphasize the significance of plasma proteomics and peptidomics in clinical applications and precision medicine.
Collapse
Affiliation(s)
- Bo He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, P. R. China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, P. R. China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, P. R. China.,Department of Pharmacology, and Provincial Key Laboratory of Pathophysiology in Ningbo University School of Medicine, Ningbo, Zhejiang, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
8
|
Cao Y, Shao L, Xin M, Zhang Y, Xu Y, Song Y, Lu H, Wang Y, Xia Y, Zhang M, Guo Y, Wang L, Qiao Z. Urine protein in patients with type I hypersensitivity is indicative of reversible renal tube injury. Life Sci 2022; 305:120735. [DOI: 10.1016/j.lfs.2022.120735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/07/2022] [Accepted: 06/23/2022] [Indexed: 11/24/2022]
|
9
|
Sun Y, Cai D, Hu W, Fang T. Identifying hub genes and miRNAs in Crohn’s disease by bioinformatics analysis. Front Genet 2022; 13:950136. [PMID: 36118873 PMCID: PMC9471261 DOI: 10.3389/fgene.2022.950136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/08/2022] [Indexed: 12/04/2022] Open
Abstract
Introduction: Crohn’s disease (CD) is a disease that manifests mainly as chronic inflammation of the gastrointestinal tract, which is still not well understood in terms of its pathogenesis. The aim of this study was to use bioinformatics analysis to identify differentially expressed genes (DEGs) and miRNAs with diagnostic and therapeutic potential in CD. Materials and methods: Three CD datasets (GSE179285, GSE102133, GSE75214) were downloaded from the Gene Expression Omnibus (GEO) database. DEGs between normal and CD tissues were identified using the GEO2R online tool. The Gene Ontology (GO) term and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the DEGs were conducted using the clusterProfiler function in the R package. Protein-protein interaction network (PPI) analysis and visualization were performed with STRING and Cytoscape. Ten hub genes were identified using cytoHubba’s MCC algorithm and validated with datasets GSE6731 and GSE52746. Finally, the miRNA gene regulatory network was constructed by Cytoscape and NetworkAnalyst to predict potential microRNAs (miRNAs) associated with DEGs. Results: A total of 97 DEGs were identified, consisting of 88 downregulated genes and 9 upregulated genes. The enriched functions and pathways of the DEGs include immune system process, response to stress, response to cytokine and extracellular region. KEGG pathway analysis indicates that the genes were significantly enriched in Cytokine-cytokine receptor interaction, IL-17 signaling pathway, Rheumatoid arthritis and TNF signaling pathway. In combination with the results of the protein-protein interaction (PPI) network and CytoHubba, 10 hub genes including IL1B, CXCL8, CXCL10, CXCL1, CXCL2, CXCL5, ICAM1, IL1RN, TIMP1 and MMP3 were selected. Based on the DEG-miRNAs network construction, 5 miRNAs including hsa-mir-21-5p, hsa-mir-93-5p, hsa-mir-98-5p, hsa-mir-1-3p and hsa-mir-335-5p were identified as potential critical miRNAs. Conclusion: In conclusion, a total of 97 DEGs, 10 hub genes and 5 miRNAs that may be involved in the progression or occurrence of CD were identified in this study, which could be regarded as biomarkers of CD.
Collapse
Affiliation(s)
- Yuxin Sun
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Daxing Cai
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Weitao Hu
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- *Correspondence: Taiyong Fang,
| |
Collapse
|
10
|
Han T, Cong H, Yu B, Shen Y. Application of peptide biomarkers in life analysis based on liquid chromatography-mass spectrometry technology. Biofactors 2022; 48:725-743. [PMID: 35816279 DOI: 10.1002/biof.1875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/18/2022] [Indexed: 12/11/2022]
Abstract
Biomedicine is developing rapidly in the 21st century. Among them, the qualitative and quantitative analysis of peptide biomarkers is of considerable importance for the diagnosis and therapy of diseases and the quality evaluation of drugs and food. The identification and quantitative analysis of peptides have been going on for decades. Traditionally, immunoassays or biological assays are generally used to quantify peptides in biological matrices. However, the selectivity and sensitivity of these methods cannot meet the requirements of the application. The separation and analysis technique of liquid chromatography-mass spectrometry (LC-MS) supplies a reliable alternative. In contrast to immunoassays, LC-MS methods are capable of providing the analytical prowess necessary to satisfy the demands of peptide biomarker research in the life sciences arena. This review article provides a historical account of the in-roads made by LC-MS technology for the detection of peptide biomarkers in the past 10 years, with the focus on the qualification/quantification developments and their applications.
Collapse
Affiliation(s)
- Tingting Han
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, China
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, China
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Hu W, Fang T, Chen X. Identification of Differentially Expressed Genes and miRNAs for Ulcerative Colitis Using Bioinformatics Analysis. Front Genet 2022; 13:914384. [PMID: 35719390 PMCID: PMC9201719 DOI: 10.3389/fgene.2022.914384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Ulcerative colitis (UC) is a chronic inflammatory disease of the intestine whose cause and underlying mechanisms are not fully understood. The aim of this study was to use bioinformatics analysis to identify differentially expressed genes (DEGs) with diagnostic and therapeutic potential in UC.Materials and methods: Three UC datasets (GSE179285, GSE75214, GSE48958) were downloaded from the Gene Expression Omnibus (GEO) database. DEGs between normal and UC tissues were identified using the GEO2R online tool. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the DEGs were performed using Metascape. Protein-protein interaction network (PPI) analysis and visualization using STRING and Cytoscape. Finally, the miRNA gene regulatory network was constructed by Cytoscape to predict potential microRNAs (miRNAs) associated with DEGs.Results: A total of 446 DEGs were identified, consisting of 309 upregulated genes and 137 downregulated genes. The enriched functions and pathways of the DEGs include extracellular matrix, regulation of cell adhesion, inflammatory response, response to cytokine, monocarboxylic acid metabolic process, response to toxic substance. The analysis of KEGG pathway indicates that the DEGs were significantly enriched in Complement and coagulation cascades, Amoebiasis, TNF signaling pathway, bile secretion, and Mineral absorption. Combining the results of the PPI network and CytoHubba, 9 hub genes including CXCL8, ICAM1, CXCR4, CD44, IL1B, MMP9, SPP1, TIMP1, and HIF1A were selected. Based on the DEG-miRNAs network construction, 7 miRNAs including miR-335-5p, mir-204-5p, miR-93-5p, miR106a-5p, miR-21-5p, miR-146a-5p, and miR-155-5p were identified as potential critical miRNAs.Conclusion: In summary, we identified DEGs that may be involved in the progression or occurrence of UC. A total of 446 DEGs,9 hub genes and 7 miRNAs were identified, which may be considered as biomarkers of UC. Further studies, however, are needed to elucidate the biological functions of these genes in UC.
Collapse
Affiliation(s)
- Weitao Hu
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiaoqing Chen
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- *Correspondence: Xiaoqing Chen,
| |
Collapse
|
12
|
Mao Z, Tan Y, Yu F, Zhao M. Discovery of NEU1 as a candidatedone. renal biomarker for proliferative lupus nephritis chronicity. Lupus Sci Med 2021; 8:8/1/e000569. [PMID: 34872988 PMCID: PMC8650488 DOI: 10.1136/lupus-2021-000569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/16/2021] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Proteomic approach was applied to identify candidate biomarkers of chronicity in patients with proliferative lupus nephritis (LN), and their clinicopathological significance and prognostic values were investigated. METHODS This study recruited 10 patients with proliferative LN and 6 normal controls (NCs) with proteomic data to compare protein expression profiles, 58 patients with proliferative LN and 10 NCs to verify proteomic data by immunohistochemistry, and 14 patients with proliferative LN with urine samples to evaluate the urinary expression of the biomarker by western blot assay. The composite endpoints included end-stage renal disease and ≥50% reduction from baseline estimated glomerular filtration rate (eGFR). RESULTS Proteomics detected 48 proteins upregulated in the group with chronicity index (CI) ≥1 compared with the CI=0 and NC groups. Further pathway analysis was enriched in 'other glycan degradation'. Neuraminidase 1 (NEU1), the most predominant protein in the pathway of other glycan degradation, was highly expressed in the kidney of patients with proliferative LN and could co-localise with podocyte, mesangial cells, endothelial cells and tubule cells. NEU1 expression in the tubulointerstitium area was significantly higher in the CI ≥1 group compared with the CI=0 and NC groups. Moreover, NEU1 expression was significantly correlated with serum creatinine value, eGFR and CI scores, respectively. Urinary NEU1 excretion in the CI ≥1 group was higher than in the CI=0 group and was also positively correlated with CI scores. Furthermore, the high expression of renal NEU1 was identified as an independent risk factor for renal prognosis by multivariate Cox regression analysis (HR, 6.462 (95% CI 1.025 to 40.732), p=0.047). CONCLUSIONS Renal NEU1 expression was associated with pathological CI scores and renal outcomes in patients with proliferative LN.
Collapse
Affiliation(s)
- Zhaomin Mao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, People's Republic of China.,Institute of Nephrology, Peking University Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China
| | - Ying Tan
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China.,Institute of Nephrology, Peking University Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China
| | - Feng Yu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China .,Institute of Nephrology, Peking University Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,Department of Nephrology, Peking University International Hospital, Beijing, People's Republic of China
| | - Minghui Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, People's Republic of China.,Institute of Nephrology, Peking University Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China
| |
Collapse
|
13
|
Identification of Immune-Related Genes in Sepsis due to Community-Acquired Pneumonia. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:8020067. [PMID: 34484417 PMCID: PMC8413041 DOI: 10.1155/2021/8020067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022]
Abstract
Background Immunosuppression has a key function in sepsis pathogenesis, so it is of great significance to find immune-related markers for the treatment of sepsis. Methods Datasets of community-acquired pneumonia (CAP) with sepsis from the ArrayExpress database were extracted. Differentially expressed genes (DEGs) between the CAP group and normal group by Limma package were performed. After calculation of immune score through the ESTIMATE algorithm, the DEGs were selected between the high immune score group and the low immune score group. Enrichment analysis of the intersected DEGs was conducted. Further, the protein-protein interaction (PPI) of the intersected DEGs was drawn by Metascape tools. Related publications of the key DEGs were searched in NCBI PubMed through Biopython models, and RT-qPCR was used to verify the expression of key genes. Results 360 intersected DEGs (157 upregulated and 203 downregulated) were obtained between the two groups. Meanwhile, the intersected DEGs were enriched in 157 immune-related terms. The PPI of the DEGs was performed, and 8 models were obtained. In sepsis-related research, eight genes were obtained with degree ≥ 10, included in the models. Conclusion CXCR3, CCR7, HLA-DMA, and GPR18 might participate in the mechanism of CAP with sepsis.
Collapse
|
14
|
Gong P, Wang M, Yang W, Chang X, Wang L, Chen F. Integrated metabolomics coupled with pattern recognition and pathway analysis to reveal molecular mechanism of cadmium-induced diabetic nephropathy. Toxicol Res (Camb) 2021; 10:777-791. [PMID: 34484669 DOI: 10.1093/toxres/tfab059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/28/2021] [Accepted: 06/06/2021] [Indexed: 12/16/2022] Open
Abstract
Diabetic nephropathy (DN) is becoming a worldwide public health problem and its pathophysiological mechanism is not well understood. Emerging evidences indicated that cadmium (Cd), an industrial material but also an environmental toxin, may be involved in the development and progression of diabetes and diabetes-related kidney disease. However, the underlying mechanism is still unclear. Herein, a DN animal model was constructed by exposing to Cd, the metabolomic profiling of DN mice were obtained by using ultra performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS), pattern recognition and pathway analysis were performed to screen potential biomarker. Moreover, western blotting was employed to verify the possible mechanism involved in the occurrence of Cd-induced DN. A total of 66 metabolites in serum have been screened out and identified as biomarkers, including free fatty acids, phospholipids, sphingomyelins, glycerides, and others. Significant differences were demonstrated between the metabolic profiles, including decreased levels of phospholipid and increased content of triglyceride, diacylglycerols, ceramide, lysophosphatidylcholine in Cd-induced DN mice compared with control. Protein expression level of p38 MAPK and Wnt/β-catenin were significantly increased. UPLC-Q-TOF/MS-based serum metabolomics coupled with pattern recognition methods and pathway analysis provide a powerful approach to identify potential biomarkers and is a new strategy to predict the underlying mechanism of disease caused by environmental toxicant.
Collapse
Affiliation(s)
- Pin Gong
- School of Food and Biotechnology, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Mengrao Wang
- School of Food and Biotechnology, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Wenjuan Yang
- School of Food and Biotechnology, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Xiangna Chang
- School of Food and Biotechnology, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Lan Wang
- School of Food and Biotechnology, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Fuxin Chen
- School of Chemistry and Chemical Engineering, Xi'an University of Science and Technology, Xi'an 710054, China
| |
Collapse
|
15
|
Wang W, Wu J, Liu P, Tang X, Pang H, Xie T, Xu F, Shao J, Chen Y, Liu B, Zheng Y. Urinary Proteomics Identifying Novel Biomarkers for the Diagnosis and Phenotyping of Carotid Artery Stenosis. Front Mol Biosci 2021; 8:714706. [PMID: 34447787 PMCID: PMC8383446 DOI: 10.3389/fmolb.2021.714706] [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: 05/25/2021] [Accepted: 07/26/2021] [Indexed: 01/12/2023] Open
Abstract
Background: Carotid artery stenosis (CAS) is caused by the formation of atherosclerotic plaques inside the arterial wall and accounts for 20–30% of all strokes. The development of an early, noninvasive diagnostic method and the identification of high-risk patients for ischemic stroke is essential to the management of CAS in clinical practice. Methods: We used the data-independent acquisition (DIA) technique to conduct a urinary proteomic study in patients with CAS and healthy controls. We identified the potential diagnosis and risk stratification biomarkers of CAS. And Ingenuity pathway analysis was used for functional annotation of differentially expressed proteins (DEPs). Furthermore, receiver operating characteristic (ROC) analysis was performed to evaluate the diagnostic values of DEPs. Results: A total of 194 DEPs were identified between CAS patients and healthy controls by DIA quantification. The bioinformatics analysis showed that these DEPs were correlated with the pathogenesis of CAS. We further identified 32 DEPs in symptomatic CAS compared to asymptomatic CAS, and biological function analysis revealed that these proteins are mainly related to immune/inflammatory pathways. Finally, a biomarker panel of six proteins (ACP2, PLD3, HLA-C, GGH, CALML3, and IL2RB) exhibited potential diagnostic value in CAS and good discriminative power for differentiating symptomatic and asymptomatic CAS with high sensitivity and specificity. Conclusions: Our study identified novel potential urinary biomarkers for noninvasive early screening and risk stratification of CAS.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianqiang Wu
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Liu
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyue Tang
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyu Pang
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Xie
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Xu
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Shao
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuexin Chen
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bao Liu
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuehong Zheng
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
He P, Zhou C, Shen H. Diagnostic value of phosphatidylethanolamine binding protein 4 levels in patients receiving nursing interventions for advanced chronic kidney disease. J Int Med Res 2021; 49:300060521996179. [PMID: 33752499 PMCID: PMC7995466 DOI: 10.1177/0300060521996179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To explore the diagnostic role of phosphatidylethanolamine binding protein 4 (PEBP4) in patients with chronic kidney disease (CKD) receiving nursing interventions. METHODS ELISA was used to evaluate serum PEBP4 levels. Receiver-operating characteristic curve analysis was used to assess diagnostic accuracy. Spearman correlation analysis was used to assess the relationships between PEBP4 levels and biochemical indexes. RESULTS Serum PEBP4 was high in CKD patients compared with healthy individuals. PEBP4 levels were positively correlated with pathological stage in CKD patients. PEBP4 had higher sensitivity for diagnosis of CKD than common indexes including blood urea nitrogen, creatinine and C-reactive protein. Among CKD patients treated with calcium channel blockers, serum PEBP4 levels declined notably and were associated with concentrations of K+, Na+, Cl- and Ca2+. Nursing interventions significantly decreased serum PEBP4 levels. A significant association between serum PEBP4 level and ionic concentration was observed in CKD patients receiving nursing interventions. CONCLUSIONS This prospective study demonstrated that PEBP4 level might represent an effective diagnostic biomarker in CKD patients. PEBP4 also acted as a valuable care compliance factor for determining the necessity for nursing interventions. Nursing interventions restored ion channel function and subsequently resulted in decreased PEBP4 levels and proteinuria.
Collapse
Affiliation(s)
- Peipei He
- Kidney Disease Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, P.R. China
- Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province Institute of Nephrology, Zhejiang University, Zhejiang, P.R. China
| | - Congli Zhou
- State Key Laboratory of Proteomics, National Center for Protein Sciences Beijing, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Huajuan Shen
- Department of Nephrology, Zhejiang Provincial People’s Hospital, Zhejiang, P.R. China
- People’s Hospital of Hangzhou Medical College, Zhejiang, P.R. China
| |
Collapse
|
17
|
Rocchetti MT, Rascio F, Castellano G, Fiorentino M, Netti GS, Spadaccino F, Ranieri E, Gallone A, Gesualdo L, Stallone G, Pontrelli P, Grandaliano G. Altered Phosphorylation of Cytoskeleton Proteins in Peripheral Blood Mononuclear Cells Characterizes Chronic Antibody-Mediated Rejection in Kidney Transplantation. Int J Mol Sci 2020; 21:ijms21186509. [PMID: 32899575 PMCID: PMC7556000 DOI: 10.3390/ijms21186509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 11/24/2022] Open
Abstract
Chronic antibody-mediated rejection (CAMR) is the major cause of kidney transplant failure. The molecular mechanisms underlying this event are still poorly defined and this lack of knowledge deeply influences the potential therapeutic strategies. The aim of our study was to analyze the phosphoproteome of peripheral blood mononuclear cells (PBMCs), to identify cellular signaling networks differentially activated in CAMR. Phosphoproteins isolated from PBMCs of biopsy proven CAMR, kidney transplant recipients with normal graft function and histology and healthy immunocompetent individuals, have been investigated by proteomic analysis. Phosphoproteomic results were confirmed by Western blot and PBMCs’ confocal microscopy analyses. Overall, 38 PBMCs samples were analyzed. A differential analysis of PBMCs’ phosphoproteomes revealed an increase of lactotransferrin, actin-related protein 2 (ARPC2) and calgranulin-B in antibody-mediated rejection patients, compared to controls. Increased expression of phosphorylated ARPC2 and its correlation to F-actin filaments were confirmed in CAMR patients. Our results are the first evidence of altered cytoskeleton organization in circulating immune cells of CAMR patients. The increased expression of phosphorylated ARPC2 found in the PBMCs of our patients, and its association with derangement of F-actin filaments, might suggest that proteins regulating actin dynamics in immune cells could be involved in the mechanism of CAMR of kidney grafts.
Collapse
Affiliation(s)
- Maria Teresa Rocchetti
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, Faculty of Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (F.R.); (G.S.N.); (F.S.); (E.R.)
| | - Federica Rascio
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, Faculty of Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (F.R.); (G.S.N.); (F.S.); (E.R.)
| | - Giuseppe Castellano
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, Faculty of Medicine University of Foggia, 71122 Foggia, Italy; (G.C.); (G.S.)
| | - Marco Fiorentino
- Nephrology Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, Faculty of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.F.); (L.G.)
| | - Giuseppe Stefano Netti
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, Faculty of Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (F.R.); (G.S.N.); (F.S.); (E.R.)
| | - Federica Spadaccino
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, Faculty of Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (F.R.); (G.S.N.); (F.S.); (E.R.)
| | - Elena Ranieri
- Clinical Pathology Unit and Center for Molecular Medicine, Department of Medical and Surgical Sciences, Faculty of Medicine, University of Foggia, 71122 Foggia, Italy; (M.T.R.); (F.R.); (G.S.N.); (F.S.); (E.R.)
| | - Anna Gallone
- Experimental Biology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, Faculty of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Loreto Gesualdo
- Nephrology Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation, Faculty of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.F.); (L.G.)
| | - Giovanni Stallone
- Nephrology Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, Faculty of Medicine University of Foggia, 71122 Foggia, Italy; (G.C.); (G.S.)
| | - Paola Pontrelli
- Experimental Biology, Department of Emergency and Organ Transplantation, Faculty of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy
- Correspondence: ; Tel.: +39-08-05-478-868
| | - Giuseppe Grandaliano
- Nephrology Unit, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy;
- Department of Translational Medicine and Surgery, Faculty of Medicine, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
18
|
Feldreich T, Nowak C, Carlsson AC, Östgren CJ, Nyström FH, Sundström J, Carrero-Roig JJ, Leppert J, Hedberg P, Giedraitis V, Lind L, Cordeiro A, Ärnlöv J. The association between plasma proteomics and incident cardiovascular disease identifies MMP-12 as a promising cardiovascular risk marker in patients with chronic kidney disease. Atherosclerosis 2020; 307:11-15. [PMID: 32702535 DOI: 10.1016/j.atherosclerosis.2020.06.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/19/2020] [Accepted: 06/18/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Previous proteomics efforts in patients with chronic kidney disease (CKD) have predominantly evaluated urinary protein levels. Therefore, our aim was to investigate the association between plasma levels of 80 cardiovascular disease-related proteins and the risk of major adverse cardiovascular events (MACE) in patients with CKD. METHODS Individuals with CKD stages 3-5 (eGFR below 60 ml min-1 [1.73 m]-2) from three community-based cohorts (PIVUS, ULSAM, SAVA), one diabetes cohort (CARDIPP) and one cohort with peripheral artery disease patients (PADVA) with information on 80 plasma protein biomarkers, assessed with a proximity extension assay, and follow-up data on incident MACE, were used as discovery sample. To validate findings and to asses generalizability to patients with CKD in clinical practice, an outpatient CKD-cohort (Malnutrition, Inflammation and Vascular Calcification (MIVC)) was used as replication sample. RESULTS In the discovery sample (total n = 1316), 249 individuals experienced MACE during 7.0 ± 2.9 years (range 0.005-12.9) of follow-up, and in the replication sample, 71 MACE events in 283 individuals over a mean ± SD change of 2.9 ± 1.2 years (range 0.1-4.0) were documented. Applying Bonferroni correction, 18 proteins were significantly associated with risk of MACE in the discovery cohort, adjusting for age and sex in order of significance, GDF-15, FGF-23, REN, FABP4, IL6, TNF-R1, AGRP, MMP-12, AM, KIM-1, TRAILR2, TNFR2, CTSL1, CSF1, PlGF, CA-125, CCL20 and PAR-1 (p < 0.000625 for all). Only matrix metalloproteinase 12 (MMP-12) was significantly associated with an increased risk of MACE in the replication sample (hazard ratio (HR) per SD increase, 1.36, 95% CI (1.07-1.75), p = 0.013). CONCLUSIONS Our proteomics analyses identified plasma MMP-12 as a promising cardiovascular risk marker in patients with CKD.
Collapse
Affiliation(s)
- Tobias Feldreich
- Education, Health and Social Studies, Dalarna University, Falun, Sweden.
| | - Christoph Nowak
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Alfred Nobels Allé 23, SE 14183, Huddinge, Sweden
| | - Axel C Carlsson
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Alfred Nobels Allé 23, SE 14183, Huddinge, Sweden
| | - Carl-Johan Östgren
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Fredrik H Nyström
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Juan-Jesus Carrero-Roig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jerzy Leppert
- Centre for Clinical Research, Uppsala University, Västerås, Sweden
| | - Pär Hedberg
- Centre for Clinical Research, Uppsala University, Västerås, Sweden
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Antonio Cordeiro
- Department of Hypertension and Nephrology, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil
| | - Johan Ärnlöv
- Education, Health and Social Studies, Dalarna University, Falun, Sweden; Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Alfred Nobels Allé 23, SE 14183, Huddinge, Sweden
| |
Collapse
|
19
|
Xie K, Kong S, Li F, Zhang Y, Wang J, Zhao W. Bioinformatics-Based Study to Investigate Potential Differentially Expressed Genes and miRNAs in Pediatric Sepsis. Med Sci Monit 2020; 26:e923881. [PMID: 32575108 PMCID: PMC7331480 DOI: 10.12659/msm.923881] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Sepsis is an extremely common health issue with a considerable mortality rate in children. Our understanding about the pathogenic mechanisms of sepsis is limited. The aim of this study was to identify the differential expression genes (DEGs) in pediatric sepsis through comprehensive analysis, and to provide specific insights for the clinical sepsis therapies in children. MATERIAL AND METHODS Three pediatric gene expression profiles (GSE25504, GSE26378, GSE26440) were downloaded from the Gene Expression Omnibus (GEO) database. The difference expression genes (DEGs) between pediatric sepsis and normal control group were screened with the GEO2R online tool. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the DEGs were performed. Cytoscape with CytoHubba were used to identify the hub genes. Finally, NetworkAnalyst was used to construct the targeted microRNAs (miRNAs) of the hub genes. RESULTS Totally, 160 overlapping upward genes and 61 downward genes were identified. In addition, 5 KEGG pathways, including hematopoietic cell lineage, Staphylococcus aureus infection, starch and sucrose metabolism, osteoclast differentiation, and tumor necrosis factor (TNF) signaling pathway, were significantly enriched using a database for labeling, visualization, and synthetic discovery. In combination with the results of the protein-protein interaction (PPI) network and CytoHubba, 9 hub genes including ITGAM, TLR8, IL1ß, MMP9, MPO, FPR2, ELANE, SPI1, and C3AR1 were selected. Combined with DEG-miRNAs visualization, 5 miRNAs, including has-miR-204-5p, has-miR-211-5p, has-miR-590-5p, and has-miR-21-5p, were predicted as possibly the key miRNAs. CONCLUSIONS Our findings will contribute to identification of potential biomarkers and novel strategies for pediatric sepsis treatment.
Collapse
Affiliation(s)
- Kexin Xie
- Laboratory Department, Dali University, Dali, Yunnan, China (mainland)
| | - Shan Kong
- Laboratory Department, Dali University, Dali, Yunnan, China (mainland)
| | - Fuxing Li
- Laboratory Department, Dali University, Dali, Yunnan, China (mainland)
| | - Yulin Zhang
- Laboratory Department, Dali University, Dali, Yunnan, China (mainland)
| | - Jing Wang
- School of Clinical Medicine, Dali University, Dali, Yunnan, China (mainland)
| | - Weidong Zhao
- Laboratory Department, Dali University, Dali, Yunnan, China (mainland)
| |
Collapse
|
20
|
Nicholas SB. Use of urinary proteomics in diagnosis and monitoring of diabetic kidney disease. Lancet Diabetes Endocrinol 2020; 8:261-262. [PMID: 32135137 DOI: 10.1016/s2213-8587(20)30067-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 01/03/2023]
Affiliation(s)
- Susanne B Nicholas
- David Geffen School of Medicine at UCLA, University of Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
21
|
Chien HJ, Xue YT, Chen HC, Wu KY, Lai CC. Proteomic analysis of rat kidney under maleic acid treatment by SWATH-MS technology. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2020; 34 Suppl 1:e8633. [PMID: 31677360 DOI: 10.1002/rcm.8633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 10/03/2019] [Accepted: 10/09/2019] [Indexed: 06/10/2023]
Abstract
RATIONALE Maleic acid is an industrial-grade chemical that is often used in adhesives, stabilizers, and preservatives. It is unknown whether long-term consumption of maleic acid modified starch is harmful to humans. However, many studies have indicated that maleic acid causes renal tubular damage in animal models, even as the associated pathways remain unclear. Sequential window acquisition of all theoretical fragment ion spectra (SWATH) is the most innovative of the label-free quantitative technologies which have better quantification performance. Therefore, SWATH technology was used to investigate the effect of maleic acid on the rat kidney proteome in this study. METHODS Sprague-Dawley(SD) rats were treated with 0 mg/kg (control), 6 mg/kg (low-dose), 10 mg/kg (medium-dose), and 60 mg/kg (high-dose) of maleic acid. After kidney protein extraction, 28% SDS-PAGE was used, followed by in-gel digestion and desalting. Next, the samples were analyzed with ultra-performance liquid chromatography (UPLC) coupled with quadrupole time-of-flight mass spectrometry (Q-TOF MS), and data-dependent acquisition (DDA) and SWATH technology were also used. The gene ontology and pathway analysis were accomplished. Ultimately, these protein biomarkers were validated by using scheduled high-resolution multiple reaction monitoring (sMRMHR ). RESULTS Comparisons of the control group with the other three groups revealed that 95, 130, and 103 proteins were expressed at significantly different levels in the control group and in the low-dose, medium-dose, and high-dose groups, respectively. According to the gene ontology analysis, the major processes that these proteins were involved in were metabolic processes, biological regulation, cellular processes, and responses to stimuli; the major functions that these proteins were involved in were binding, hydrolase activity, catalytic activity, and oxidoreductase activity; and the major cellular components hat they were involved in were the cytoplasm, extracellular region, membrane, and mitochondria. According to the KEGG pathway analysis, these proteins were involved in 35 pathways, five of which, the carbohydrate metabolism, folate biosynthesis, renal tubular resorption, amino acid metabolism, and Ras signaling pathways, are discussed in this study. Ultimately, 19 proteins involved in 12 important pathways were validated by sMRMHR . CONCLUSIONS It was demonstrated that maleic acid caused insufficient energy production, which might lead to a decrease in the activity of the sodium-potassium ATP pump and hydrogen ion ATP pump, which could in turn have caused renal tubular resorption and hydrogen ion regulation to be blocked, thus leading to the accumulation of hydrogen ions in the renal tubules, which would then result in renal tubular acidification followed finally by Fanconi syndrome.
Collapse
Affiliation(s)
- Han-Ju Chien
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Yu-Ting Xue
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Hsin-Chang Chen
- Institute of Occupational Medicine and Industrial Hygiene, College of Public Health, National Taiwan University, No. 17, ShiuJou Road, Taipei, 10055, Taiwan
| | - Kuen-Yuh Wu
- Institute of Occupational Medicine and Industrial Hygiene, College of Public Health, National Taiwan University, No. 17, ShiuJou Road, Taipei, 10055, Taiwan
| | - Chien-Chen Lai
- Institute of Molecular Biology, National Chung Hsing University, Taichung, 40227, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, 40447, Taiwan
- Advanced Plant Biotechnology Center, National Chung Hsing University, Taichung, 40402, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei City, 11490, Taiwan
| |
Collapse
|
22
|
Cao C, Xiao Z, Ge C, Wu Y. Application and Research Progress of Proteomics in Chicken Meat Quality and Identification: A Review. FOOD REVIEWS INTERNATIONAL 2020. [DOI: 10.1080/87559129.2020.1733594] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Changwei Cao
- College of Food Science, Sichuan Agricultural University, Ya’ An, Sichuan, China
| | - Zhichao Xiao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Changrong Ge
- Yunnan Agricultural University, Kunming, Yunnan, China
| | - Yinglong Wu
- College of Food Science, Sichuan Agricultural University, Ya’ An, Sichuan, China
| |
Collapse
|
23
|
Sirolli V, Pieroni L, Di Liberato L, Urbani A, Bonomini M. Urinary Peptidomic Biomarkers in Kidney Diseases. Int J Mol Sci 2019; 21:E96. [PMID: 31877774 PMCID: PMC6982248 DOI: 10.3390/ijms21010096] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 12/20/2022] Open
Abstract
In order to effectively develop personalized medicine for kidney diseases we urgently need to develop highly accurate biomarkers for use in the clinic, since current biomarkers of kidney damage (changes in serum creatinine and/or urine albumin excretion) apply to a later stage of disease, lack accuracy, and are not connected with molecular pathophysiology. Analysis of urine peptide content (urinary peptidomics) has emerged as one of the most attractive areas in disease biomarker discovery. Urinary peptidome analysis allows the detection of short and long-term physiological or pathological changes occurring within the kidney. Urinary peptidomics has been applied extensively for several years now in renal patients, and may greatly improve kidney disease management by supporting earlier and more accurate detection, prognostic assessment, and prediction of response to treatment. It also promises better understanding of kidney disease pathophysiology, and has been proposed as a "liquid biopsy" to discriminate various types of renal disorders. Furthermore, proteins being the major drug targets, peptidome analysis may allow one to evaluate the effects of therapies at the protein signaling pathway level. We here review the most recent findings on urinary peptidomics in the setting of the most common kidney diseases.
Collapse
Affiliation(s)
- Vittorio Sirolli
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS.Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy; (V.S.); (L.D.L.)
| | - Luisa Pieroni
- Proteomics and Metabonomics Unit, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| | - Lorenzo Di Liberato
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS.Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy; (V.S.); (L.D.L.)
| | - Andrea Urbani
- Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Department of Laboratory Diagnostic and Infectious Diseases, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Mario Bonomini
- Nephrology and Dialysis Unit, Department of Medicine, G. d’Annunzio University, Chieti-Pescara, SS.Annunziata Hospital, Via dei Vestini, 66013 Chieti, Italy; (V.S.); (L.D.L.)
| |
Collapse
|
24
|
Small molecules from natural products targeting the Wnt/β-catenin pathway as a therapeutic strategy. Biomed Pharmacother 2019; 117:108990. [DOI: 10.1016/j.biopha.2019.108990] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/09/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
|
25
|
Chronic kidney disease: Biomarker diagnosis to therapeutic targets. Clin Chim Acta 2019; 499:54-63. [PMID: 31476302 DOI: 10.1016/j.cca.2019.08.030] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD), characterized as renal dysfunction, is recognized as a major public health problem with high morbidity and mortality worldwide. Unfortunately, there are no obvious clinical symptoms in early stage disease until severe damage has occurred. Further complicating early diagnosis and treatment is the lack of sensitive and specific biomarkers. As such, novel biomarkers are urgently needed. Metabolomics has shown an increasing potential for identifying underlying disease mechanisms, facilitating clinical diagnosis and developing pharmaceutical treatments for CKD. Recent advances in metabolomics revealed that CKD was closely associated with the dysregulation of numerous metabolites, such as amino acids, lipids, nucleotides and glycoses, that might be exploited as potential biomarkers. In this review, we summarize recent metabolomic applications based on animal model studies and in patients with CKD and highlight several biomarkers that may play important roles in diagnosis, intervention and development of new therapeutic strategies.
Collapse
|
26
|
Banerjee S, Wong ACY, Yan X, Wu B, Zhao H, Tibshirani RJ, Zare RN, Brooks JD. Early detection of unilateral ureteral obstruction by desorption electrospray ionization mass spectrometry. Sci Rep 2019; 9:11007. [PMID: 31358807 PMCID: PMC6662848 DOI: 10.1038/s41598-019-47396-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/16/2019] [Indexed: 01/08/2023] Open
Abstract
Desorption electrospray ionization mass spectrometry (DESI-MS) is an emerging analytical tool for rapid in situ assessment of metabolomic profiles on tissue sections without tissue pretreatment or labeling. We applied DESI-MS to identify candidate metabolic biomarkers associated with kidney injury at the early stage. DESI-MS was performed on sections of kidneys from 80 mice over a time course following unilateral ureteral obstruction (UUO) and compared to sham controls. A predictive model of renal damage was constructed using the LASSO (least absolute shrinkage and selection operator) method. Levels of lipid and small metabolites were significantly altered and glycerophospholipids comprised a significant fraction of altered species. These changes correlate with altered expression of lipid metabolic genes, with most genes showing decreased expression. However, rapid upregulation of PG(22:6/22:6) level appeared to be a hitherto unknown feature of the metabolic shift observed in UUO. Using LASSO and SAM (significance analysis of microarrays), we identified a set of well-measured metabolites that accurately predicted UUO-induced renal damage that was detectable by 12 h after UUO, prior to apparent histological changes. Thus, DESI-MS could serve as a useful adjunct to histology in identifying renal damage and demonstrates early and broad changes in membrane associated lipids.
Collapse
Affiliation(s)
- Shibdas Banerjee
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA.,Department of Chemistry, Indian Institute of Science Education and Research Tirupati, Tirupati, 517507, India
| | - Anny Chuu-Yun Wong
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xin Yan
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Bo Wu
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hongjuan Zhao
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Robert J Tibshirani
- Departments of Biomedical Data Sciences, and of Statistics, Stanford University, Stanford, CA, 94305, USA
| | - Richard N Zare
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA.
| | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
27
|
Biancotto G, Bovo D, Mastrorilli E, Manuali E, Angeletti R, Stella R. TMT-Based Proteomics Profiling of Bovine Liver Underscores Protein Markers of Anabolic Treatments. Proteomics 2019; 19:e1800422. [PMID: 30865377 DOI: 10.1002/pmic.201800422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/22/2019] [Indexed: 12/23/2022]
Abstract
Illegal use of growth promoter compounds in food production exposes consumers to health risk. Surveillance of such practices is based on direct detection of drugs or related metabolites by HPLC-MS/MS. Screening strategies focusing on indirect biological responses are considered promising tools to improve surveillance. In this study, an untargeted shotgun proteomics approach based on tandem mass tags (TMTs) is carried out to identify proteins altered in bovine liver after different anabolic treatments. Three controlled pharmacological treatments with dexamethasone, a combination of dexamethasone and clenbuterol, or a combination of sexual steroids (trenbolone and estradiol) are analyzed. Untargeted TMT analysis of liver digests by high resolution MS allowed for the relative quantification of proteins. Thanks to partial least squarediscriminant analysis, a set of proteins capable to classify animals treated with dexamethasone alone (11 proteins), or in combination with clenbuterol (13 proteins) are identified. No significant difference is found upon administration of sexual steroids. After relative quantification of candidate markers by parallel reaction monitoring (PRM), two predictive models are trained to validate protein markers. Finally, an independent animal set of control bulls and bulls treated with dexamethasone is analyzed by PRM to further validate a predictive model giving an accuracy of 100%.
Collapse
Affiliation(s)
- Giancarlo Biancotto
- Department of Chemistry, Istituto Zooprofilattico Sperimentale delle Venezie, 35020, Legnaro PD, Italy
| | - Davide Bovo
- Department of Chemistry, Istituto Zooprofilattico Sperimentale delle Venezie, 35020, Legnaro PD, Italy
| | - Eleonora Mastrorilli
- Department of Food Safety, Istituto Zooprofilattico Sperimentale delle Venezie, 35020, Legnaro PD, Italy
| | - Elisabetta Manuali
- Istituto Zooprofilattico Sperimentale dell'Umbria e delle Marche "Togo Rosati", 06126, Perugia, Italy
| | - Roberto Angeletti
- Department of Chemistry, Istituto Zooprofilattico Sperimentale delle Venezie, 35020, Legnaro PD, Italy
| | - Roberto Stella
- Department of Chemistry, Istituto Zooprofilattico Sperimentale delle Venezie, 35020, Legnaro PD, Italy
| |
Collapse
|
28
|
Liang L, Li Y, Tian X, Zhou J, Zhong L. Comprehensive lipidomic, metabolomic and proteomic profiling reveals the role of immune system in vitiligo. Clin Exp Dermatol 2019; 44:e216-e223. [PMID: 30859585 DOI: 10.1111/ced.13961] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Vitiligo is a common depigmentation disorder resulting from destruction of melanocytes, and has both genetic and environmental influences. Although genomic analyses have been performed to investigate the pathogenesis of vitiligo, the lipidomics, metabolomics and proteomics of serum have not been reported, and the role of small molecules and serum proteins in vitiligo remains unknown. AIM To study the metabolite and protein profiles in patients with vitiligo and healthy controls (HCs). METHODS Plasma samples from 60 participants (29 patients with vitiligo and 31 HCs) were analysed. Untargeted lipidomics, metabolomics and isobaric tags for relative and absolute quantification-based proteomics were performed using high performance liquid chromatography-tandem mass spectrometry. In addition, to validate differentially expressed metabolites in patients with vitiligo, plasma enzyme-linked immunosorbent assay was performed. RESULTS We identified differential expression of several metabolites and proteins involved in the immune system. Among these metabolites and proteins, lysophosphatidylcholine, platelet-activating factor, sn-glycerol-3-phosphocholine, succinic acid, CXCL4 and CXCL7 were significantly elevated in the plasma of patients with vitiligo, while aspartate was downregulated. CONCLUSION Our study has characterized several serum metabolites and proteins that could be potential candidate biomarkers in vitiligo, and provides a comprehensive insight into the role of immune system and aspartate metabolism in vitiligo.
Collapse
Affiliation(s)
- L Liang
- Department of Biophysics and, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Y Li
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - X Tian
- Jilin People's Hospital, Jilin, China
| | - J Zhou
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,Beijing Advanced Innovation Center for Big Data-based Precision Medicine, Beihang University, Beijing, 100191, China
| | - L Zhong
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing, China
| |
Collapse
|
29
|
Unilateral ureteral obstruction causes gut microbial dysbiosis and metabolome disorders contributing to tubulointerstitial fibrosis. Exp Mol Med 2019; 51:1-18. [PMID: 30918245 PMCID: PMC6437207 DOI: 10.1038/s12276-019-0234-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/11/2018] [Accepted: 12/20/2018] [Indexed: 12/18/2022] Open
Abstract
Chronic kidney disease (CKD) increases the risk and prevalence of cardiovascular disease (CVD) morbidity and mortality. Recent studies have revealed marked changes in the composition of the microbiome and the metabolome and their potential influence in renal disease and CVD via the accumulation of microbial-derived uremic toxins. However, the effect of unilateral ureteral obstruction (UUO) on the gut microbiome and circulating metabolites is unknown. Male Sprague-Dawley rats were randomized to UUO and sham-operated control groups. Renal histology, colonic microbiota, and plasma metabolites were examined two weeks later. We employed 16S rRNA sequence and untargeted metabolomic analyses to explore the changes in colonic microbiota and plasma metabolites and their relationship with tubulointerstitial fibrosis (TIF). The UUO rats exhibited tubular atrophy and dilatation, interstitial fibrosis and inflammatory cell infiltration in the obstructed kidney. UUO rats showed significant colonic enrichment and depletion of genera. Significant differences were identified in 219 plasma metabolites involved in lipid, amino acid, and bile acid metabolism, which were consistent with gut microbiota-related metabolism. Interestingly, tryptophan and its metabolites kynurenine, 5-hydroxytryptophan and 5-hydroxytryptamine levels, which were linked with TIF, correlated with nine specific genera. Plasma tryptophan level was positively correlated with Clostridium IV, Turicibacter, Pseudomonas and Lactobacillales, and negatively correlated with Oscillibacter, Blautia, and Intestinimonas, which possess the genes encoding tryptophan synthase (K16187), indoleamine 2,3-dioxygenase (K00463) and tryptophan 2,3-dioxygenase (K00453) and their corresponding enzymes (EC:1.13.11.52 and EC:1.13.11.11) that exacerbate TIF. In conclusion, UUO results in profound changes in the gut microbiome and circulating metabolites, events that contribute to the pathogenesis of inflammation and TIF.
Collapse
|
30
|
AKF-PD alleviates diabetic nephropathy via blocking the RAGE/AGEs/NOX and PKC/NOX Pathways. Sci Rep 2019; 9:4407. [PMID: 30867431 PMCID: PMC6416244 DOI: 10.1038/s41598-018-36344-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022] Open
Abstract
Diabetic nephropathy (DN) is a major complication of diabetes. Currently, drugs are not available to effectively control the disease. Fluorofenidone (AKF-PD) is a recently developed drug; it possesses activities in reducing DN progression in preclinical research. Nonetheless, its renal protection and the underlying mechanisms have not been thoroughly investigated. We report here that AKF-PD significantly alleviatesrenal oxidative stress (OS) in db/dbmice through downregulation of Nicotinamide Adenine Dinucleotide Phosphate (NADPH) oxidase and upregulation of glutathione peroxidase and superoxide dismutase, thereby protecting kidney from DN pathogenesis. AKF-PD likely reduces OS through the advanced glycation end products (AGE) and protein kinase C (PKC) pathways. While renal AGEs, PKCα, PKCβ, and NADPH oxidase 4 (NOX4) were all substantially upregulated in db/db mice compared to db/m animals, AKF-PD robustly downregulated all these events to the basal levelsdetected in db/m mice. In primary human renal mesangial cells (HMCs), high glucose (HG) elevated receptor for advanced glycation endproducts (RAGE), PKCα, PKCβ and NOX4 activity, and induced the production of reactive oxygen species (ROS); these events were all inhibited by AKF-PD. Furthermore, HG led to mitochondrial damagein HMCs;AKF-PD conferred protection on the damage. Knockdown of either PKCα or PKCβ reduced HG-induced ROS production and mitochondrial damage in HMCs. The knockdown significantly enhanced AKF-PD-mediated inhibition of ROS production and mitochondrial damage in HG-treated HMCs. Collectively, our study demonstrates that AKF-PD protects renal function under diabetes conditions in part through inhibition of OS during DN pathogenesis. AKF-PD can be explored for clinical applications in DN therapy.
Collapse
|
31
|
Wu HY, Bi R, Sun T, Xie F. Deletion of Dicer blocks osteogenic differentiation via the inhibition of Wnt signalling. Mol Med Rep 2019; 19:2897-2905. [PMID: 30816532 DOI: 10.3892/mmr.2019.9941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 01/29/2019] [Indexed: 11/06/2022] Open
Abstract
Micro (mi)RNAs are small, non‑coding RNAs and have been reported to have important roles in the epigenetic control of bone development. miRNAs markedly regulate osteoblast differentiation through stages of maturation as well as the activities of osteogenic signaling pathways. Dicer is an important endoribonuclease that regulates miRNA maturation. Previous studies have demonstrated that Dicer deletion decreases fetal survival and bone formation, while excision in differentiated osteoblasts increases bone mass. However, the underlying molecular mechanisms remain unclear. In the present study, whether the deletion of Dicer affects Wnt signaling, which exhibits important roles during osteogenesis, was investigated. Bone marrow stromal cells (BMSCs) were used as an osteogenic model. Dynamic changes of seven Wnt genes and downstream T‑cell factor 1 (Tcf‑1)/lymphoid enhancing binding factor were observed during the osteogenic differentiation of BMSCs, which revealed different roles at early and late differentiation stages. Following the stable knockdown of Dicer in BMSCs using lentiviral short hairpin RNA, osteogenic differentiation was blocked, and the levels of important osteogenic differentiation markers (runt related transcription factor 2 and alkaline phosphatase) were markedly inhibited. Furthermore, stage specific regulation of Wnt genes in Dicer‑deficient BMSCs was investigated in the present study. At the early differentiation stage (days 5‑7), knockdown of Dicer led to the inhibition of Wnt1, Wnt7 and Wnt10b, as well as the upregulation of Wnt4, Wnt10a and Tcf‑1. At the late stage of differentiation (days 14‑21), knockdown of Dicer significantly suppressed the expression levels of all of the included Wnt genes as well as Tcf‑1, with the exception of Wnt10a. The upregulation of Wnt10a following the deletion of Dicer was maintained throughout all stages of differentiation. In addition, differential regulation of Wnt genes and Tcf‑1 were revealed to be associated with dynamic changes in their expression levels during osteogenic differentiation. Furthermore, the four putative Wnt10a‑targeting miRNAs were investigated in the present study, and the results demonstrated that they were upregulated during osteogenic differentiation, which suggested that inhibition of Wnt10a may be an important factor associated with osteogenic differentiation. In conclusion, the present study investigated the mechanism underlying the regulation of Wnt signalling by Dicer during osteogenesis, and identified potential miRNAs targeting the components of Wnt signalling influenced by Dicer. Collectively, the present study identified the association between Dicer and Wnt signalling during bone development.
Collapse
Affiliation(s)
- Hong-Yan Wu
- Pharmacy Department, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Rui Bi
- Pharmacy Department, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| | - Ting Sun
- Department of Clinical Pharmacology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Fei Xie
- Pharmacy Department, Tangshan People's Hospital, Tangshan, Hebei 063001, P.R. China
| |
Collapse
|
32
|
Boulangé CL, Rood IM, Posma JM, Lindon JC, Holmes E, Wetzels JFM, Deegens JKJ, Kaluarachchi MR. NMR and MS urinary metabolic phenotyping in kidney diseases is fit-for-purpose in the presence of a protease inhibitor. Mol Omics 2019; 15:39-49. [DOI: 10.1039/c8mo00190a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
When using an appropriate data analysis pipeline, protease inhibitor (PI)-containing urine samples are fit-for-purpose for metabolic phenotyping of patients with nephrotic syndrome and proteinuria.
Collapse
Affiliation(s)
| | - Ilse M. Rood
- Department of Nephrology
- Radboud University Medical Center
- Nijmegen
- The Netherlands
| | - Joram M. Posma
- Imperial College London
- Division of Computational and Systems Medicine
- Department of Surgery and Cancer
- Faculty of Medicine
- London SW7 2AZ
| | - John C. Lindon
- Metabometrix Ltd
- London SW7 2AZ
- UK
- Imperial College London
- Division of Computational and Systems Medicine
| | - Elaine Holmes
- Metabometrix Ltd
- London SW7 2AZ
- UK
- Imperial College London
- Division of Computational and Systems Medicine
| | - Jack F. M. Wetzels
- Department of Nephrology
- Radboud University Medical Center
- Nijmegen
- The Netherlands
| | - Jeroen K. J. Deegens
- Department of Nephrology
- Radboud University Medical Center
- Nijmegen
- The Netherlands
| | | |
Collapse
|
33
|
Chen DQ, Hu HH, Wang YN, Feng YL, Cao G, Zhao YY. Natural products for the prevention and treatment of kidney disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 50:50-60. [PMID: 30466992 DOI: 10.1016/j.phymed.2018.09.182] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/18/2018] [Accepted: 09/17/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) is one of the common causes resulting in a high morbidity and mortality. Renal fibrosis is the main pathological features of CKD. Natural products have begun to gain widely popularity worldwide for promoting healthcare and preventing CKD, and have been used as a conventional or complementary therapy for CKD treatment. PURPOSE The present paper reviewed the therapeutic effects of natural products on CKD and revealed the molecular mechanisms of their anti-fibrosis. METHODS All the available information on natural products against renal fibrosis was collected via a library and electronic search (using Web of Science, Pubmed, ScienceDirect, Splinker, etc.). RESULTS Accumulated evidence demonstrated that natural products exhibited the beneficial effects for CKD treatment and against renal fibrosis. This review presents an overview of the molecular mechanism of CKD and natural products against renal fibrosis, followed by an in-depth discussion of their molecular mechanism of natural products including isolated compounds and crude extracts against renal fibrosis in vitro and in vivo. A number of isolated compounds have been confirmed to retard renal fibrosis. CONCLUSION The review provides comprehensive insights into pathophysiological mechanisms of CKD and natural products against renal fibrosis. Particular challenges are presented and placed within the context of future applications of natural products against renal fibrosis.
Collapse
Affiliation(s)
- Dan-Qian Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - He-He Hu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Yan-Ni Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Ya-Long Feng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang 310053, China
| | - Ying-Yong Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
34
|
Yinchenhao Decoction Alleviates Liver Fibrosis by Regulating Bile Acid Metabolism and TGF-β/Smad/ERK Signalling Pathway. Sci Rep 2018; 8:15367. [PMID: 30337590 PMCID: PMC6194075 DOI: 10.1038/s41598-018-33669-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/31/2018] [Indexed: 12/12/2022] Open
Abstract
Yinchenhao decoction (YCHD), comprising Yinchenhao (Artemisiae Scopariae Herba), Zhizi (Gardeniae Fructus) and Dahuang (Radix Rhei et Rhizoma), is widely used for treating various diseases. We aimed to investigate the bile acid metabolic mechanism of YCHD in dimethylnitrosamine (DMN)-induced liver fibrosis model. Rats received DMN (10 mg/kg, intraperitoneally) for four successive weeks for liver fibrosis induction and were treated with YCHD for the last 2 weeks. Histopathological analysis showed that YCHD prevented DMN-induced histopathological changes in liver tissues. Serum liver function in YCHD group improved. Ultraperformance liquid chromatography-mass spectrometry analysis showed that YCHD significantly restored both free and conjugated bile acid levels increased by DMN, to normal levels. RT-qPCR results showed that YCHD treatment upregulated the expression of genes related to bile acid synthesis, reabsorption, and excretion. Western blotting analysis showed that YCHD downregulated α-SMA, TGF-β1, p-Smad3, and p-ERK1/2 expression in chenodeoxycholic acid (CDCA)-activated hepatic stellate cells (HSCs). The viability of CDCA-activated HSCs significantly increased after treatment with YCHD and PD98059 (an ERK inhibitor) compared to YCHD treatment alone. Our findings suggest that YCHD alleviated DMN-induced liver fibrosis by regulating enzymes responsible for bile acid metabolism. Additionally, it inhibits CDCA-induced HSC proliferation and activation via TGF-β1/Smad/ERK signalling pathway.
Collapse
|
35
|
Validation of a Novel Modified Aptamer-Based Array Proteomic Platform in Patients with End-Stage Renal Disease. Diagnostics (Basel) 2018; 8:diagnostics8040071. [PMID: 30297602 PMCID: PMC6316431 DOI: 10.3390/diagnostics8040071] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/24/2018] [Accepted: 10/02/2018] [Indexed: 12/30/2022] Open
Abstract
End stage renal disease (ESRD) is characterized by complex metabolic abnormalities, yet the clinical relevance of specific biomarkers remains unclear. The development of multiplex diagnostic platforms is creating opportunities to develop novel diagnostic and therapeutic approaches. SOMAscan is an innovative multiplex proteomic platform which can measure >1300 proteins. In the present study, we performed SOMAscan analysis of plasma samples and validated the measurements by comparison with selected biomarkers. We compared concentrations of SOMAscan-measured prostate specific antigen (PSA) between males and females, and validated SOMAscan concentrations of fibroblast growth factor 23 (FGF23), FGF receptor 1 (FGFR1), and FGFR4 using Enzyme-Linked immunosorbent assay (ELISA). The median (25th and 75th percentile) SOMAscan PSA level in males and females was 4304.7 (1815.4 to 7259.5) and 547.8 (521.8 to 993.4) relative fluorescence units (p = 0.002), respectively, suggesting biological plausibility. Pearson correlation between SOMAscan and ELISA was high for FGF23 (R = 0.95, p < 0.001) and FGFR4 (R = 0.69, p < 0.001), indicating significant positive correlation, while a weak correlation was found for FGFR1 (R = 0.13, p = 0.16). In conclusion, there is a good to near-perfect correlation between SOMAscan and standard immunoassays for FGF23 and FGFR4, but not for FGFR1. This technology may be useful to simultaneously measure a large number of plasma proteins in ESRD, and identify clinically important prognostic markers to predict outcomes.
Collapse
|
36
|
Yu X, Xia Y, Zeng L, Zhang X, Chen L, Yan S, Zhang R, Zhao C, Zeng Z, Shu Y, Huang S, Lei J, Yuan C, Zhang L, Feng Y, Liu W, Huang B, Zhang B, Luo W, Wang X, Zhang H, Haydon RC, Luu HH, He TC, Gan H. A blockade of PI3Kγ signaling effectively mitigates angiotensin II-induced renal injury and fibrosis in a mouse model. Sci Rep 2018; 8:10988. [PMID: 30030497 PMCID: PMC6054654 DOI: 10.1038/s41598-018-29417-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/06/2018] [Indexed: 12/27/2022] Open
Abstract
Chronic kidney disease (CKD) poses a formidable challenge for public healthcare worldwide as vast majority of patients with CKD are also at risk of accelerated cardiovascular disease and death. Renal fibrosis is the common manifestation of CKD that usually leads to end-stage renal disease although the molecular events leading to chronic renal fibrosis and eventually chronic renal failure remain to be fully understood. Nonetheless, emerging evidence suggests that an aberrant activation of PI3Kγ signaling may play an important role in regulating profibrotic phenotypes. Here, we investigate whether a blockade of PI3Kγ signaling exerts any beneficial effect on alleviating kidney injury and renal fibrosis. Using a mouse model of angiotensin II (Ang II)-induced renal damage, we demonstrate that PI3Kγ inhibitor AS605240 effectively mitigates Ang II-induced increases in serum creatinine and blood urea nitrogen, renal interstitial collagen deposition, the accumulation of ECM proteins and the expression of α-Sma and fibrosis-related genes in vivo. Mechanistically, we reveal that AS605240 effectively inhibits Ang II-induced cell proliferation and phosphorylation of Akt in fibroblast cells. Furthermore, we demonstrate that Ang II-upregulated expression of IL-6, Tnf-α, IL-1β and Tgf-β1 is significantly attenuated in the mice treated with AS605240. Taken together, our results demonstrate that PI3Kγ may function as a critical mediator of Ang II-induced renal injury and fibrosis. It is thus conceivable that targeted inhibition of PI3Kγ signaling may constitute a novel therapeutic approach to the clinical management of renal fibrosis, renal hypertension and/or CKD.
Collapse
Affiliation(s)
- Xinyi Yu
- Departments of Nephrology, Orthopaedic Surgery, Cardiology, General Surgery, and Clinical Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yunfeng Xia
- Departments of Nephrology, Orthopaedic Surgery, Cardiology, General Surgery, and Clinical Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liyi Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Infection Control, Zhuzhou Central Hospital, and the Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, China
| | - Xi Zhang
- Departments of Nephrology, Orthopaedic Surgery, Cardiology, General Surgery, and Clinical Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Liqun Chen
- Departments of Nephrology, Orthopaedic Surgery, Cardiology, General Surgery, and Clinical Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Shujuan Yan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | - Ruyi Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | - Chen Zhao
- Departments of Nephrology, Orthopaedic Surgery, Cardiology, General Surgery, and Clinical Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | - Shifeng Huang
- Departments of Nephrology, Orthopaedic Surgery, Cardiology, General Surgery, and Clinical Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Jiayan Lei
- Departments of Nephrology, Orthopaedic Surgery, Cardiology, General Surgery, and Clinical Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Chengfu Yuan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Biochemistry and Molecular Biology, China Three Gorges University School of Medicine, Yichang, 443002, China
| | - Linghuan Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | - Yixiao Feng
- Departments of Nephrology, Orthopaedic Surgery, Cardiology, General Surgery, and Clinical Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Wei Liu
- Departments of Nephrology, Orthopaedic Surgery, Cardiology, General Surgery, and Clinical Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Bo Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Infection Control, Zhuzhou Central Hospital, and the Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, China
- Department of Clinical Laboratory Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, China
| | - Bo Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Key Laboratory of Orthopaedic Surgery of Gansu Province and the Department of Orthopaedic Surgery, the Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Wenping Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Xi Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | - Hongmei Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, 401147, China
| | - Rex C Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Hue H Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA.
| | - Hua Gan
- Departments of Nephrology, Orthopaedic Surgery, Cardiology, General Surgery, and Clinical Laboratory Medicine, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
37
|
Hu HH, Chen DQ, Wang YN, Feng YL, Cao G, Vaziri ND, Zhao YY. New insights into TGF-β/Smad signaling in tissue fibrosis. Chem Biol Interact 2018; 292:76-83. [PMID: 30017632 DOI: 10.1016/j.cbi.2018.07.008] [Citation(s) in RCA: 701] [Impact Index Per Article: 100.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 07/01/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023]
Abstract
Transforming growth factor-β1 (TGF-β1) is considered as a crucial mediator in tissue fibrosis and causes tissue scarring largely by activating its downstream small mother against decapentaplegic (Smad) signaling. Different TGF-β signalings play different roles in fibrogenesis. TGF-β1 directly activates Smad signaling which triggers pro-fibrotic gene overexpression. Excessive studies have demonstrated that dysregulation of TGF-β1/Smad pathway was an important pathogenic mechanism in tissue fibrosis. Smad2 and Smad3 are the two major downstream regulator that promote TGF-β1-mediated tissue fibrosis, while Smad7 serves as a negative feedback regulator of TGF-β1/Smad pathway thereby protects against TGF-β1-mediated fibrosis. This review presents an overview of the molecular mechanisms of TGF-β/Smad signaling pathway in renal, hepatic, pulmonary and cardiac fibrosis, followed by an in-depth discussion of their molecular mechanisms of intervention effects both in vitro and in vivo. The role of TGF-β/Smad signaling pathway in tumor or cancer is also discussed. Additionally, the current advances also highlight targeting TGF-β/Smad signaling pathway for the prevention of tissue fibrosis. The review reveals comprehensive pathophysiological mechanisms of tissue fibrosis. Particular challenges are presented and placed within the context of future applications against tissue fibrosis.
Collapse
Affiliation(s)
- He-He Hu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Dan-Qian Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Yan-Ni Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Ya-Long Feng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, Irvine, CA, 92897, USA
| | - Ying-Yong Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China.
| |
Collapse
|
38
|
Ma X, He L. The intervention effect of zuogui pill on chronic kidney disease-mineral and bone disorder regulatory factor. Biomed Pharmacother 2018; 106:54-60. [PMID: 29957466 DOI: 10.1016/j.biopha.2018.06.092] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 06/15/2018] [Accepted: 06/15/2018] [Indexed: 12/29/2022] Open
Abstract
Chronic kidney disease-mineral and bone disorder (CKD-MBD) play a critical role in the pathogenesis of cardiovascular complications in patients with chronic kidney disease (CKD). Zuogui pill as a traditional Chinese herbal drug has been used for nourish kidney essence improve bone malnutrition of renal bone disease by regulating the metabolism of calcium and phosphorus and participating in osteoblast metabolism. In the present study, 5/6 nephrectomy rat model was used to reveal the mechanism of zuogui pill in treatment of CKD-MBD. Compared with sham rats, the levels of serum phosphorus, PTH, iPTH and creatinine were significantly decreased, while the serum calcium level was significantly increased, and the Cbfa1 protein level was significantly decreased and FGF23 protein level was significantly increased by Zuogui pill treatment. Compared with model rats, the BMD of rat was significantly increased by Zuogui pill treatment. Histological analysis revealed that the kidney injury of rats with CKD was significantly reduced by zuogui pill treatment. Compared with model rats, the CYP27B1 mRNA level was significantly increased, and the PTH mRNA level and NaPiIIa protein level were significantly decreased in the kidney by zuogui pill treatment. We inferred that zuogui pill exhibited potential therapeutic effects on CKD-MBD in the rats by regulating bone metabolism and nourish kidney.
Collapse
Affiliation(s)
- Xiaohong Ma
- Chenxinghai Hospital of Zhongshan City, No. 18 Zhuyuan Road, Xiaolan Town, Zhongshan, Guangdong, 528415, China.
| | - Liqun He
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine and Pharmacology, No. 185 Pu'an Road, Huangpu District, Shanghai, 200021, China
| |
Collapse
|
39
|
Cai X, Wang L, Wang X, Hou F. Silence of IGFBP7 suppresses apoptosis and epithelial mesenchymal transformation of high glucose induced-podocytes. Exp Ther Med 2018; 16:1095-1102. [PMID: 30112052 PMCID: PMC6090473 DOI: 10.3892/etm.2018.6298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/16/2018] [Indexed: 12/15/2022] Open
Abstract
Insulin-like growth factor-binding protein 7 (IGFBP7) has been identified as a secreted protein associated with a number of cellular processes. However, the specific regulatory mechanisms of IGFBP7 on podocytes of diabetic nephropathy (DN) are yet to be elucidated. In the present study, podocytes were identified initially via an immunofluorescence assay using an anti-synaptopodin antibody. It was subsequently demonstrated that glucose promoted podocyte proliferation in a time- and dose-dependent manner via MTT assay. In addition, IGFBP7 expression was silenced in podocytes via siRNA, the effects of which were evaluated using western blotting and reverse transcription-quantitative polymerase chain reaction. It was demonstrated that silencing IGFBP7 inhibited apoptosis and epithelial mesenchymal transformation (EMT) of podocytes mediated by high glucose (HG). Transforming growth factor (TGF)-β1/mothers against decapentaplegic homolog (Smad) signaling was associated with proliferation, apoptotic activities and EMT. Therefore, the expression levels of TGF-β1/Smad pathway were detected, and it was observed that silencing IGFBP7 suppressed the TGF-β1/Smad pathway in podocytes induced by HG. These findings suggested that IGFBP7 may serve as a potential therapeutic target for DN.
Collapse
Affiliation(s)
- Xiaojun Cai
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang 150036, P.R. China
| | - Lei Wang
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang 150036, P.R. China
| | - Xuling Wang
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang 150036, P.R. China
| | - Fengyan Hou
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang 150036, P.R. China
| |
Collapse
|