1
|
Bahaabadi ZJ, Javid-Naderi MJ, Kesharwani P, Karav S, Sahebkar A. A Review on Biosensors for Quantification of MCP-1 as a Potential Biomarker in Diseases. Immunology 2025. [PMID: 40365864 DOI: 10.1111/imm.13944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/11/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1) as a chemokine is essential for inflammation-related processes. It regulates immunological responses and cell migration, which contribute to inflammation. Many disorders are exacerbated by this chemokine, which attracts or grows other inflammatory cells, including monocytes/macrophages, at the site of infection or tissue injury. The elevated concentrations of MCP-1 are associated with the pathogenesis of many diseases, such as cancer, cardiovascular disease, kidney disease, and neuroinflammatory disease. Therefore, monitoring this inflammatory biomarker in the body has been recommended and strongly advised to make an accurate diagnosis and prognosis. Although MCP-1 is of great importance in disease processes, few biosensing approaches are specifically designed to detect this molecule. These are often electrochemical and optical techniques. Rapid and accurate diagnosis of inflammatory diseases by identifying biomarkers has had a great effect on the advancement of biosensors. Improved biosensor technology expansion prevents excessive prices and low sensitivity, enabling quick and correct diagnosis and tracking of disease processes. This review will concentrate on the biological functions of MCP-1, its significance in different disorders, and the features and applications of biosensors designed for MCP-1 detection and quantification.
Collapse
Affiliation(s)
- Zahra Jamalizadeh Bahaabadi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Javad Javid-Naderi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh, India
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Ma J, Xu X, Zhang Y, Guo X, Sun Y, Wang X, Zhao L, Shen Q. Pulsed Radiofrequency Alleviates Acute Soft Tissue Injury in Rats by Regulating the TNF/mTOR Signaling Pathway. Photobiomodul Photomed Laser Surg 2025; 43:198-206. [PMID: 40197902 DOI: 10.1089/photob.2024.0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025] Open
Abstract
Objective: Acute traumatic muscle injuries are common and result in substantial loss of time and risk of recurrence. Pulsed radiofrequency (PR) is a strategy that has been gradually adopted for treating muscle injuries in clinical practice. However, the molecular mechanism underlying its therapeutic effects is currently unclear. Materials and Methods: In this study, we screened the gene expression profiles of rats with muscle contusion obtained from the online dataset GSE162565. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses of the differentially expressed genes were conducted. Further, we established an acute soft tissue injury (ASTI) rat model and applied PR treatment. Muscle swelling rate analysis, malondialdehyde (MAD) and superoxide dismutase (SOD) content, inflammatory cytokine release, and hematoxylin and eosin staining of the gastrocnemius muscles of ASTI and ASTI + PR rats were performed, and the results were compared with those of control rats. Further, we evaluated the gene expression of Ccl1, interleukin-6 (IL-6), nuclear factor-kappa-B-inhibitor alpha (Nfkbia), Akt1, Jun, Fos, and Caps3 in the model and PR-treated groups, all of which are key genes in the tumor necrosis factor (TNF)/mechanistic target of rapamycin (mTOR) signaling pathway according to the KEGG analysis. Results: The results revealed that 52 genes involved in the TNF/mTOR signaling pathway were closely associated with ASTI progression in rats. PR treatment significantly reduced the malondialdehyde content but increased the SOD content in ASTI model rat muscles, efficiently alleviated muscle contusions and reduced TNF-α and IL-1β production. Moreover, PR treatment significantly decreased Ccl1, IL-6, and Nfkbia expression but increased Akt1, Jun, Fos, and Caps3 levels in ASTI models. These data indicate that PR alleviated ASTI in rats by mediating redox homeostasis and the inflammatory response, which might be modulated by the TNF/mTOR signaling pathway. Conclusions: Thus, this study contributes to the understanding of ASTI progression and provides more substantial information about the genetic mechanism underlying the therapeutic effects of PR on ASTI.
Collapse
Affiliation(s)
- Jianyun Ma
- Department of Pain Treatment, The People's Hospital of Suzhou New District, Suzhou City, China
| | - Xue Xu
- Medical Research Center, The People's Hospital of Suzhou New District, Suzhou City, China
| | - Ying Zhang
- Department of Pain Treatment, The People's Hospital of Suzhou New District, Suzhou City, China
| | - Xiaoli Guo
- Department of Pain Treatment, The People's Hospital of Suzhou New District, Suzhou City, China
| | - Yunzhong Sun
- Department of Pain Treatment, The People's Hospital of Suzhou New District, Suzhou City, China
| | - Xiaochuan Wang
- Department of Pain Treatment, The People's Hospital of Suzhou New District, Suzhou City, China
| | - Lei Zhao
- Department of Pain Treatment, The People's Hospital of Suzhou New District, Suzhou City, China
| | - Qiming Shen
- Department of Pain Treatment, The People's Hospital of Suzhou New District, Suzhou City, China
| |
Collapse
|
3
|
Peng Z, Pang H, Wu H, Peng X, Tan Q, Lin S, Wei B. CCL2 promotes proliferation, migration and angiogenesis through the MAPK/ERK1/2/MMP9, PI3K/AKT, Wnt/β‑catenin signaling pathways in HUVECs. Exp Ther Med 2022; 25:77. [PMID: 36684650 PMCID: PMC9842938 DOI: 10.3892/etm.2022.11776] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/28/2022] [Indexed: 12/28/2022] Open
Abstract
Severe bone trauma can lead to poor or delayed bone healing and nonunion. Bone regeneration is based on the interaction between osteogenesis and angiogenesis. Angiogenesis serves a unique role in the repair and remodeling of bone defects. Monocyte chemoattractant protein-1, also known as CC motif ligand 2 (CCL2), is a member of the CC motif chemokine family and was the first human chemokine to be revealed to be an effective chemokine of monocytes. However, its underlying mechanism in angiogenesis of bone defect repair remains to be elucidated. Therefore, the present study investigated the detailed mechanism by which CCL2 promoted angiogenesis in bone defects based on cell and animal model experiments. In the present study, CCL2 promoted proliferation, migration and tube formation in human umbilical vein endothelial cells (HUVECs) in a concentration-dependent manner. Western blot analysis revealed that treatment of HUVECs with CCL2 upregulated the protein expression levels of rho-associated coiled-coil-containing protein kinase (Rock)1, Rock2, N-cadherin, c-Myc and VEGFR2. Furthermore, CCL2 promoted the expression of MAPK/ERK1/2/MMP9, PI3K/AKT and Wnt/β-catenin signaling pathway-related proteins, which also demonstrated that CCL2 promoted these functions in HUVECs. Immunohistochemical staining of Sprague Dawley rat femurs following bone defects revealed that VEGF expression was positive in the newly formed bone area in each group, while the expression area of VEGF in the CCL2 addition group was markedly increased. Therefore, CCL2 is a potential therapeutic approach for bone defect repair and reconstruction through the mechanism of angiogenesis-osteogenesis coupling.
Collapse
Affiliation(s)
- Zhonghua Peng
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - He Pang
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Hang Wu
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xin Peng
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Qichao Tan
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Sien Lin
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China,Correspondence to: Dr Sien Lin or Dr Bo Wei, Department of Orthopedics Center, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Xiashan, Zhanjiang, Guangdong 524001, P.R. China
| | - Bo Wei
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China,Correspondence to: Dr Sien Lin or Dr Bo Wei, Department of Orthopedics Center, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Xiashan, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
4
|
Bayly-Jones C, Lupton CJ, Fritz C, Venugopal H, Ramsbeck D, Wermann M, Jäger C, de Marco A, Schilling S, Schlenzig D, Whisstock JC. Helical ultrastructure of the metalloprotease meprin α in complex with a small molecule inhibitor. Nat Commun 2022; 13:6178. [PMID: 36261433 PMCID: PMC9581967 DOI: 10.1038/s41467-022-33893-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
The zinc-dependent metalloprotease meprin α is predominantly expressed in the brush border membrane of proximal tubules in the kidney and enterocytes in the small intestine and colon. In normal tissue homeostasis meprin α performs key roles in inflammation, immunity, and extracellular matrix remodelling. Dysregulated meprin α is associated with acute kidney injury, sepsis, urinary tract infection, metastatic colorectal carcinoma, and inflammatory bowel disease. Accordingly, meprin α is the target of drug discovery programs. In contrast to meprin β, meprin α is secreted into the extracellular space, whereupon it oligomerises to form giant assemblies and is the largest extracellular protease identified to date (~6 MDa). Here, using cryo-electron microscopy, we determine the high-resolution structure of the zymogen and mature form of meprin α, as well as the structure of the active form in complex with a prototype small molecule inhibitor and human fetuin-B. Our data reveal that meprin α forms a giant, flexible, left-handed helical assembly of roughly 22 nm in diameter. We find that oligomerisation improves proteolytic and thermal stability but does not impact substrate specificity or enzymatic activity. Furthermore, structural comparison with meprin β reveal unique features of the active site of meprin α, and helical assembly more broadly.
Collapse
Affiliation(s)
- Charles Bayly-Jones
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia
| | - Christopher J Lupton
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia
| | - Claudia Fritz
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | - Hariprasad Venugopal
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, 3800, VIC, Australia
| | - Daniel Ramsbeck
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | - Michael Wermann
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
| | | | - Alex de Marco
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia
| | - Stephan Schilling
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany
- Hochschule Anhalt, University of Applied Sciences, Köthen, Germany
| | - Dagmar Schlenzig
- Department for Drug Design and Target Validation (IZI-MWT), Fraunhofer Institute for Cell Therapy and Immunology, Halle, Germany.
| | - James C Whisstock
- Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia.
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC, Australia.
- EMBL Australia, Monash University, Melbourne, VIC, 3800, Australia.
- ACRF Department of Cancer Biology and Therapeutics, John Curtin School of Medical Research, Australian National University, Canberra, ACT, 2601, Australia.
| |
Collapse
|
5
|
Rahn S, Becker-Pauly C. Meprin and ADAM proteases as triggers of systemic inflammation in sepsis. FEBS Lett 2022; 596:534-556. [PMID: 34762736 DOI: 10.1002/1873-3468.14225] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/19/2021] [Accepted: 10/28/2021] [Indexed: 12/24/2022]
Abstract
Systemic inflammatory disorders (SIDs) comprise a broad range of diseases characterized by dysregulated excessive innate immune responses. Severe forms of SIDs can lead to organ failure and death, and their increasing incidence represents a major issue for the healthcare system. Protease-mediated ectodomain shedding of cytokines and their receptors represents a central mechanism in the regulation of inflammatory responses. The metalloprotease A disintegrin and metalloproteinase (ADAM) 17 is the best-characterized ectodomain sheddase capable of releasing TNF-α and soluble IL-6 receptor, which are decisive factors of systemic inflammation. Recently, meprin metalloproteases were also identified as IL-6 receptor sheddases and activators of the pro-inflammatory cytokines IL-1β and IL-18. In different mouse models of SID, particularly those mimicking a sepsis-like phenotype, ADAM17 and meprins have been found to promote disease progression. In this review, we summarize the role of ADAM10, ADAM17, and meprins in the onset and progression of sepsis and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Sascha Rahn
- Biochemical Institute, Christian-Albrechts-University Kiel, Germany
| | | |
Collapse
|
6
|
Berner DK, Wessolowski L, Armbrust F, Schneppenheim J, Schlepckow K, Koudelka T, Scharfenberg F, Lucius R, Tholey A, Kleinberger G, Haass C, Arnold P, Becker‐Pauly C. Meprin β cleaves TREM2 and controls its phagocytic activity on macrophages. FASEB J 2020; 34:6675-6687. [DOI: 10.1096/fj.201902183r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/03/2019] [Accepted: 03/12/2020] [Indexed: 11/11/2022]
Affiliation(s)
| | - Luisa Wessolowski
- Unit for Degradomics of the Protease Web, Biochemical Institute University of Kiel Kiel Germany
| | - Fred Armbrust
- Unit for Degradomics of the Protease Web, Biochemical Institute University of Kiel Kiel Germany
| | | | - Kai Schlepckow
- German Center for Neurodegenerative Diseases (DZNE) Munich Germany
| | - Tomas Koudelka
- Systematic Proteomics & Bioanalytics Institute for Experimental Medicine University of Kiel Kiel Germany
| | - Franka Scharfenberg
- Unit for Degradomics of the Protease Web, Biochemical Institute University of Kiel Kiel Germany
| | - Ralph Lucius
- Anatomical Institute University of Kiel Kiel Germany
| | - Andreas Tholey
- Systematic Proteomics & Bioanalytics Institute for Experimental Medicine University of Kiel Kiel Germany
| | - Gernot Kleinberger
- Biomedical Center, Biochemistry Ludwig‐Maximilians‐Universität Munich Munich Germany
- Munich Cluster for Systems Neurology Munich Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich Germany
- Biomedical Center, Biochemistry Ludwig‐Maximilians‐Universität Munich Munich Germany
- Munich Cluster for Systems Neurology Munich Germany
| | | | - Christoph Becker‐Pauly
- Unit for Degradomics of the Protease Web, Biochemical Institute University of Kiel Kiel Germany
| |
Collapse
|
7
|
Ahmed F, Mwiza JM, Fernander M, Yahaya I, Abousaad S, Ongeri EM. Meprin-β activity modulates the β-catalytic subunit of protein kinase A in ischemia-reperfusion-induced acute kidney injury. Am J Physiol Renal Physiol 2020; 318:F1147-F1159. [PMID: 32174142 DOI: 10.1152/ajprenal.00571.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Meprin metalloproteases have been implicated in the progression of kidney injury. Previous work from our group has shown that meprins proteolytically process the catalytic subunit of protein kinase A (PKA-C), resulting in decreased PKA-C kinase activity. The goal of the present study was to determine the PKA-C isoforms impacted by meprin-β and whether meprin-β expression affects downstream mediators of the PKA signaling pathway in ischemia-reperfusion (IR)-induced kidney injury. IR was induced in 12-wk-old male wild-type (WT) and meprin-β knockout (βKO) mice. Madin-Darby canine kidney cells transfected with meprin-β cDNA were also subjected to 2 h of hypoxia. Western blot analysis was used to evaluate levels of total PKA-C, PKA-Cα, PKA-Cβ, phosphorylated (p-)PKA-C, and p-ERK1/2. Meprin-β expression enhanced kidney injury as indicated by levels of neutrophil gelatinase-associated lipocalin and cystatin C. IR-associated decreases were observed in levels of p-PKA-C in kidney tissue from WT mice but not βKO mice, suggesting that meprin-β expression/activity is responsible for the in vivo reduction in kinase activity. Significant increases in levels of PKA-Cβ were observed in kidney lysates for WT mice but not βKO mice at 6 h post-IR. Proximal tubule PKA-Cβ increases in WT but not βKO kidneys were demonstrated by fluorescent microscopy. Furthermore, IR-induced injury was associated with significant increases in p-ERK levels for both genotypes. The present data demonstrate that meprin-β enhances IR-induced kidney injury in part by modulating mediators of the PKA-Cβ signaling pathway.
Collapse
Affiliation(s)
- Faihaa Ahmed
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| | - Jean-Marie Mwiza
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| | - Mizpha Fernander
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| | - Ismaila Yahaya
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| | - Shaymaa Abousaad
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| | - Elimelda Moige Ongeri
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| |
Collapse
|
8
|
Chen Y, Xu D, Yao J, Wei Z, Li S, Gao X, Cai W, Mao N, Jin F, Li Y, Zhu Y, Li S, Liu H, Yang F, Xu H. Inhibition of miR-155-5p Exerts Anti-Fibrotic Effects in Silicotic Mice by Regulating Meprin α. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 19:350-360. [PMID: 31877411 PMCID: PMC6939030 DOI: 10.1016/j.omtn.2019.11.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/06/2019] [Accepted: 11/17/2019] [Indexed: 02/07/2023]
Abstract
Silicosis is a fatal profession-related disease linked to long-term inhalation of silica. The present study aimed to determine whether meprin α, a master regulator of anti-fibrotic peptide N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP), is diminished by miR-155-5p in silicotic and control lung macrophages and fibroblasts upon activation. NR8383 macrophages, primary lung fibroblasts, and mouse embryonic fibroblasts were used to evaluate the expression and function of meprin α and miR-155-5p. In vitro meprin α manipulation was performed by recombinant mouse meprin α protein, actinonin (its inhibitor), and small interfering RNA knockdown. Macrophage and fibroblast activation was assessed by western blotting, real-time PCR, matrix deposition, and immunohistochemical staining. The roles of meprin α and miR-155-5p were also investigated in mice exposed to silica. We found that the meprin α level was stably repressed in silicotic rats. In vitro, silica decreased meprin α, and exogenous meprin α reduced activation of macrophages and fibroblasts induced by profibrotic factors. miR-155-5p negatively regulated Mep1a by binding to the 3′ untranslated region. Treatment with anti-miR-155-5p elevated meprin α, ameliorated macrophage and fibroblast activation, and attenuated lung fibrosis in mice induced by silica. The sustained repression of meprin α and beneficial effects of its rescue by inhibition of miR-155-5p during silicosis indicate that miR-155-5p/meprin α are two of the major regulators of silicosis.
Collapse
Affiliation(s)
- Yingying Chen
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Dingjie Xu
- Traditional Chinese Medicine College, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Jingxin Yao
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Zhongqiu Wei
- Basic Medical College, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Shifeng Li
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Xuemin Gao
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Wenchen Cai
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Na Mao
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Fuyu Jin
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Yaqian Li
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Ying Zhu
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Shumin Li
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Heliang Liu
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, Hebei 063210, China; School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Fang Yang
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, Hebei 063210, China; School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Hong Xu
- Medical Research Center, Hebei Key Laboratory for Organ Fibrosis Research, North China University of Science and Technology, Tangshan, Hebei 063210, China.
| |
Collapse
|
9
|
Gooding J, Cao L, Ahmed F, Mwiza JM, Fernander M, Whitaker C, Acuff Z, McRitchie S, Sumner S, Ongeri EM. LC-MS-based metabolomics analysis to identify meprin-β-associated changes in kidney tissue from mice with STZ-induced type 1 diabetes and diabetic kidney injury. Am J Physiol Renal Physiol 2019; 317:F1034-F1046. [PMID: 31411076 PMCID: PMC6843037 DOI: 10.1152/ajprenal.00166.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 08/04/2019] [Accepted: 08/06/2019] [Indexed: 01/22/2023] Open
Abstract
Meprin metalloproteases have been implicated in the pathophysiology of diabetic kidney disease (DKD). Single-nucleotide polymorphisms in the meprin-β gene have been associated with DKD in Pima Indians, a Native American ethnic group with an extremely high prevalence of DKD. In African American men with diabetes, urinary meprin excretion positively correlated with the severity of kidney injury. In mice, meprin activity decreased at the onset of diabetic kidney injury. Several studies have identified meprin targets in the kidney. However, it is not known how proteolytic processing of the targets by meprins impacts the metabolite milieu in kidneys. In the present study, global metabolomics analysis identified differentiating metabolites in kidney tissues from wild-type and meprin-β knockout mice with streptozotocin (STZ)-induced type 1 diabetes. Kidney tissues were harvested at 8 wk post-STZ and analyzed by hydrophilic interaction liquid chromatography ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry. Principal component analysis identified >200 peaks associated with diabetes. Meprin expression-associated metabolites with strong variable importance of projection scores were indoxyl sulfate, N-γ-l-glutamyl-l-aspartic acid, N-methyl-4-pyridone-3-carboxamide, inosine, and cis-5-decenedioic acid. N-methyl-4-pyridone-3-carboxamide has been previously implicated in kidney injury, and its isomers, 4-PY and 2-PY, are markers of peroxisome proliferation and inflammation that correlate with creatinine clearance and glucose tolerance. Meprin deficiency-associated differentiating metabolites with high variable importance of projection scores were cortisol, hydroxymethoxyphenylcarboxylic acid-O-sulfate, and isovaleryalanine. The data suggest that meprin-β activity enhances diabetic kidney injury in part by altering the metabolite balance in kidneys, favoring high levels of uremic toxins such as indoxyl sulfate and N-methyl-pyridone-carboxamide.
Collapse
Affiliation(s)
- Jessica Gooding
- National Institutes of Health Common Fund Eastern Regional Comprehensive Metabolomics Resource Core, RTI International, Research Park, North Carolina
| | - Lei Cao
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| | - Faihaa Ahmed
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| | - Jean-Marie Mwiza
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| | - Mizpha Fernander
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| | - Courtney Whitaker
- National Institutes of Health Common Fund Eastern Regional Comprehensive Metabolomics Resource Core, RTI International, Research Park, North Carolina
| | - Zach Acuff
- National Institutes of Health Common Fund Eastern Regional Comprehensive Metabolomics Resource Core, RTI International, Research Park, North Carolina
| | - Susan McRitchie
- National Institutes of Health Common Fund Eastern Regional Comprehensive Metabolomics Resource Core, RTI International, Research Park, North Carolina
- Department of Nutrition, School of Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - Susan Sumner
- National Institutes of Health Common Fund Eastern Regional Comprehensive Metabolomics Resource Core, RTI International, Research Park, North Carolina
- Department of Nutrition, School of Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - Elimelda Moige Ongeri
- Department of Biology, North Carolina A&T State University, Greensboro, North Carolina
| |
Collapse
|
10
|
Meprin β metalloproteases associated with differential metabolite profiles in the plasma and urine of mice with type 1 diabetes and diabetic nephropathy. BMC Nephrol 2019; 20:141. [PMID: 31023251 PMCID: PMC6485094 DOI: 10.1186/s12882-019-1313-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/27/2019] [Indexed: 02/07/2023] Open
Abstract
Background Meprin metalloproteases are abundantly expressed in the brush border membranes of kidney proximal tubules and small intestines. Meprins are also expressed in podocytes and leukocytes (monocytes and macrophages). Meprins are implicated in the pathophysiology of diabetic nephropathy (DN) but underlying mechanisms are not fully understood. Single nucleotide polymophisms (SNPs) in the meprin β gene were associated with DKD in human subjects. Furthermore, meprin α and β double deficiency resulted in more severe kidney injury and higher mortality rates in mice with Streptozotocin (STZ)-induced type 1 diabetes. Identification of meprin substrates has provided insights on how meprins could modulate kidney injury. Meprin targets in the kidney include extracellular matrix (ECM) proteins, modulators of inflammation, and proteins involved in the protein kinase A (PKA) and PKC signaling pathways. The current study used a global metabolomics approach to determine how meprin β expression impacts the metabolite milieu in diabetes and DKD. Methods Low dose STZ was used to induce type 1 diabetes in 8-week old wild-type (WT) and meprin β knockout (βKO) mice. Blood and urine samples were obtained at 4 and 8 weeks post-STZ injection. Assays for albumin, creatinine, neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule − 1 (KIM-1), and cystatin C were used for biochemical assessment of kidney injury. Data for biomarkers of kidney injury utilized two-way ANOVA. Metabolomics data analysis utilized UPLC-QTOF MS and multivariate statistics. Results The number of metabolites with diabetes-associated changes in levels were significantly higher in the WT mice when compared to meprin βKO counterparts. Annotated meprin β expression-associated metabolites with strong variable importance in projection (VIP) scores play roles in lipid metabolism (LysoPC(16:1(9Z)), taurocholic acid), amino acid metabolism (indoxyl sulfate, hippuric acid), and neurotransmitter/stress hormone synthesis (cortisol, 3-methoxy-4-hydroxyphenylethylene glycolsulfate, homovanillic acid sulfate). Metabolites that associated with meprin β deficiency include; 3,5-dihydroxy-3′,4′-dimethoxy-6,7-methylenedioxyflavone 3-glucuronide, pantothenic acid, and indoxyl glucuronide (all decreased in plasma). Conclusion Taken together, the annotated metabolites suggest that meprin β impacts complications of diabetes such as DKD by altering distinct metabolite profiles. Electronic supplementary material The online version of this article (10.1186/s12882-019-1313-2) contains supplementary material, which is available to authorized users.
Collapse
|
11
|
Deng Y, Luan S, Zhang Q, Xiao Y. Retracted: Long noncoding RNA THRIL contributes in lipopolysaccharide-induced HK-2 cells injury by sponging miR-34a. J Cell Biochem 2019; 120:1444-1456. [PMID: 30414207 DOI: 10.1002/jcb.27354] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 06/22/2018] [Indexed: 02/02/2023]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with unknown etiology. Nowadays, several long noncoding RNAs (lncRNAs) have been reported as molecular alterations involved in SLE. This study aimed to reveal the function of TNF-related and HNRNPL-related immunoregulatory lncRNA (THRIL) in SLE. Human epithelial HK-2 cells were exposed to lipopolysaccharide (LPS) to mimic an in vitro SLE model. Then, the functions of THRIL, miR-34a, and monocyte chemoattractant protein-1 (MCP-1), as well as their correlations were detected. LncRNA THRIL was highly expressed in the LPS-stimulated cells, and THRIL overexpression aggravated LPS-induced cell damage as cell viability was decreased, and apoptosis and the release of proinflammatory cytokines were increased. THRIL worked as a sponge of microRNA-34a (miR-34a) and it could directly target MCP-1. Furthermore, MCP-1-activated JNK and Wnt/β-catenin signaling pathways. In conclusion, this study suggested that lncRNA THRIL might be a key regulator participating in LPS-induced injury in HK-2 cells. THRIL overexpression aggravated LPS-induced injury possibly via sponging miR-34a, and thus preventing MCP-1 from degradation by miR-34a. The THRIL/miR-34a/MCP-1 axis might play critical roles in SLE.
Collapse
Affiliation(s)
- Yao Deng
- Department of Rheumatology and Immunology, Shengli Oilfield Central Hospital, Dongying, China
| | - Sen Luan
- Department of Nephrology, Shengli Oilfield Central Hospital, Dongying, China
| | - Qi Zhang
- Department of Rheumatology and Immunology, Shengli Oilfield Central Hospital, Dongying, China
| | - Ying Xiao
- Department of Nephrology, Shengli Oilfield Central Hospital, Dongying, China
| |
Collapse
|
12
|
Herzog C, Haun RS, Kaushal GP. Role of meprin metalloproteinases in cytokine processing and inflammation. Cytokine 2018; 114:18-25. [PMID: 30580156 DOI: 10.1016/j.cyto.2018.11.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/16/2018] [Accepted: 11/25/2018] [Indexed: 11/15/2022]
Abstract
Meprin metalloendopeptidases, comprising α and β isoforms, are widely expressed in mammalian cells and organs including kidney, intestines, lungs, skin, and bladder, and in a variety of immune cells and cancer cells. Meprins proteolytically process many inflammatory mediators, including cytokines, chemokines, and other bioactive proteins and peptides that control the function of immune cells. The knowledge of meprin-mediated processing of inflammatory mediators and other target substrates provides a pathophysiologic link for the involvement of meprins in the pathogenesis of many inflammatory disorders. Meprins are now known to play important roles in inflammatory diseases including acute kidney injury, sepsis, urinary tract infections, bladder inflammation, and inflammatory bowel disease. The proteolysis of epithelial and endothelial barriers including cell junctional proteins by meprins promotes leukocyte influx into areas of tissue damage to result in inflammation. Meprins degrade extracellular matrix proteins; this ability of meprins is implicated in the cell migration of leukocytes and the invasion of tumor cells that express meprins. Proteolytic processing and maturation of procollagens provides evidence that meprins are involved in collagen maturation and deposition in the fibrotic processes involved in the formation of keloids and hypertrophic scars and lung fibrosis. This review highlights recent progress in understanding the role of meprins in inflammatory disorders in both human and mouse models.
Collapse
Affiliation(s)
- Christian Herzog
- Central Arkansas Veterans Healthcare System and University of Arkansas for Medical Sciences, Department of Internal Medicine, Little Rock, AR, USA
| | - Randy S Haun
- Central Arkansas Veterans Healthcare System and University of Arkansas for Medical Sciences, Department of Pharmaceutical Sciences, Little Rock, AR, USA
| | - Gur P Kaushal
- Central Arkansas Veterans Healthcare System and University of Arkansas for Medical Sciences, Department of Internal Medicine, Little Rock, AR, USA; Central Arkansas Veterans Healthcare System and University of Arkansas for Medical Sciences, Department of Biochemistry, Little Rock, AR, USA.
| |
Collapse
|
13
|
Undiagnosed Kidney Injury in Uninsured and Underinsured Diabetic African American Men and Putative Role of Meprin Metalloproteases in Diabetic Nephropathy. Int J Nephrol 2018; 2018:6753489. [PMID: 29854459 PMCID: PMC5949186 DOI: 10.1155/2018/6753489] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 02/23/2018] [Accepted: 03/05/2018] [Indexed: 11/18/2022] Open
Abstract
Diabetes is the leading cause of chronic kidney disease. African Americans are disproportionately burdened by diabetic kidney disease (DKD) and end stage renal disease (ESRD). Disparities in DKD have genetic and socioeconomic components, yet its prevalence in African Americans is not adequately studied. The current study used multiple biomarkers of DKD to evaluate undiagnosed DKD in uninsured and underinsured African American men in Greensboro, North Carolina. Participants consisted of three groups: nondiabetic controls, diabetic patients without known kidney disease, and diabetic patients with diagnosed DKD. Our data reveal undiagnosed kidney injury in a significant proportion of the diabetic patients, based on levels of both plasma and urinary biomarkers of kidney injury, namely, urinary albumin to creatinine ratio, kidney injury molecule-1, cystatin C, and neutrophil gelatinase-associated lipocalin. We also found that the urinary levels of meprin A, meprin B, and two kidney meprin targets (nidogen-1 and monocytes chemoattractant protein-1) increased with severity of kidney injury, suggesting a potential role for meprin metalloproteases in the pathophysiology of DKD in this subpopulation. The study also demonstrates a need for more aggressive tests to assess kidney injury in uninsured diabetic patients to facilitate early diagnosis and targeted interventions that could slow progression to ESRD.
Collapse
|