1
|
Shanahati D, Yang T, Fang L, Nijiati Y, Aihemaitituoheti A, Abudukeremu A, Aikemu A. 18β-glycyrrhetinic acid improves pulmonary hypertension by regulating the vascular non-inflammatory molecule-1/L-arginine/nitric oxide signaling pathway. Eur J Pharmacol 2025; 996:177382. [PMID: 39988095 DOI: 10.1016/j.ejphar.2025.177382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
PURPOSE Pulmonary hypertension (PH) is a progressive cardiopulmonary disease characterized by elevated pulmonary arterial pressure and resistance. 18β-Glycyrrhetinic acid (18β-GA), a major bioactive component in Glycyrrhiza glabra L., exhibits various pharmacological activities. Although 18β-GA has been found to alleviate PH, the underlying mechanisms are still unclear. This study was aimed to elucidate the molecular processes by which 18β-GA attenuates PH. PATIENTS AND METHODS A monocrotaline-induced PH rat model was created, and cardiac function and pulmonary arterial pressure were assessed. Rat blood markers were analyzed via enzyme-linked immunosorbent assay, and protein levels of Vanin-1, NF-κB, IL-6, TNF-α, PPAR-γ, and iNOS in lung tissue were determined by western blotting. A hypoxia-induced pulmonary artery smooth muscle cell (PASMCs) proliferation model was created, and cell proliferation and migration were evaluated using CCK-8 and scratch wound assays. Cell cycle, apoptosis, and mitochondrial membrane potential were assessed via flow cytometry, and protein levels were measured using western blotting. RESULTS PH rats treated with, 18β-GA exhibited reduced mean pulmonary arterial pressure (mPAP) and delayed pulmonary vascular remodeling. In lung tissue, Vanin-1, NF-κB, IL-6, and TNF-α were downregulated, while PPAR-γ and iNOS were upregulated. PASMCs treated with 18β-GA exhibited reduced hypoxia-induced proliferation and increased apoptosis. In addition, NO levels in the supernatant of PASMCs were elevated, and Vanin-1 and NF-κB were downregulated, whereas PPAR-γ and iNOS were upregulated. CONCLUSION Our findings reveal a novel mechanism by which 18β-GA attenuates PH by improving pulmonary vascular remodeling and inhibiting PASMCs proliferation via the Vanin-1/L-Arg/NO pathway.
Collapse
MESH Headings
- Animals
- Glycyrrhetinic Acid/analogs & derivatives
- Glycyrrhetinic Acid/pharmacology
- Glycyrrhetinic Acid/therapeutic use
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Signal Transduction/drug effects
- Male
- Rats
- Cell Proliferation/drug effects
- Nitric Oxide/metabolism
- Pulmonary Artery/drug effects
- Pulmonary Artery/pathology
- Rats, Sprague-Dawley
- Humans
- Cell Movement/drug effects
- Apoptosis/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
Collapse
Affiliation(s)
- Daliya Shanahati
- College of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Tao Yang
- Central Laboratory, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Lei Fang
- College of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | - Yiliyaer Nijiati
- Central Laboratory, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China
| | | | | | - Ainiwaer Aikemu
- College of Pharmacy, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China; Department of Pharmacy, Xinjiang Hetian College, Hetian, Xinjiang, 848000, China; Xinjiang Key Laboratory of Hetian Characteristic Chinese Traditional Medicine Research, Hetian, Xinjiang, 848000, China; Engineering Research Center for Quality Control of Uyghur Medicinal Materials and Preparations, Hetian, Xinjiang Uyghur Autonomous Region, 848000, China.
| |
Collapse
|
2
|
Tretiak S, Mendes Maia T, Rijsselaere T, Van Immerseel F, Ducatelle R, Impens F, Antonissen G. Comprehensive analysis of blood proteome response to necrotic enteritis in broiler chicken. Vet Res 2025; 56:88. [PMID: 40275387 PMCID: PMC12023520 DOI: 10.1186/s13567-025-01519-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/14/2025] [Indexed: 04/26/2025] Open
Abstract
Necrotic enteritis (NE) in broiler chickens is caused by the overgrowth of toxin-producing strains of Clostridium (C.) perfringens. This study aims to analyze the blood proteome of broiler chickens affected by NE, providing insights into the host's response to the infection. Using MS/MS-based proteomics, blood plasma samples from broilers with necrotic lesions of different severity were analyzed and compared to healthy controls. A total of 412 proteins were identified, with 63 showing significant differences; for 25 of those correlation with disease severity was observed. Functional analysis revealed that proteins affected by NE were predominantly associated with the immune and signaling processes and extracellular matrix (ECM) structures. Notably, regulated proteins were significantly involved in bioprocesses related to complement activation, acute phase reaction, proteolysis and humoral immune response. The proteomics findings suggest that the changes in plasma proteins in response to NE are driven by the host's intensified efforts to counteract the infection, demonstrating a.o. activation of ECM-degrading proteases (MMP2, TIMP2), acute phase response (HPS5, CP, EXFABP, TF, VNN) and notable reduction in basement membrane (BM) and ECM-related peptides (PLOD2, POSTN, COL1A1/2, HSPG2, NID2) detected in the blood of NE-affected birds. Moreover, the findings underscore a coordinated effort of the host to mitigate the C. perfringens infection via activating immune (a.o., C3, CFH, MASP2, MBL2) and acute phase (CP, ORM, TF, ExFAB) related proteins. This study provides a deeper understanding of the host-pathogen interactions and identifies potential biomarkers and targets for therapeutic intervention. Data are available via ProteomeXchange with identifier PXD054172.
Collapse
Affiliation(s)
- Svitlana Tretiak
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Livestock Gut Health Team (LiGHT) Ghent, Ghent University, 9820, Merelbeke, Belgium
- Impextraco NV, Wiekevorstsesteenweg 38, 2220, Heist-op-den-Berg, Belgium
| | - Teresa Mendes Maia
- VIB-UGent Center for Medical Biotechnology, VIB, 9052, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9052, Ghent, Belgium
- VIB Proteomics Core, VIB, 9052, Ghent, Belgium
| | - Tom Rijsselaere
- Impextraco NV, Wiekevorstsesteenweg 38, 2220, Heist-op-den-Berg, Belgium
| | - Filip Van Immerseel
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Livestock Gut Health Team (LiGHT) Ghent, Ghent University, 9820, Merelbeke, Belgium
| | - Richard Ducatelle
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Livestock Gut Health Team (LiGHT) Ghent, Ghent University, 9820, Merelbeke, Belgium
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, 9052, Ghent, Belgium.
- Department of Biomolecular Medicine, Ghent University, 9052, Ghent, Belgium.
- VIB Proteomics Core, VIB, 9052, Ghent, Belgium.
| | - Gunther Antonissen
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Livestock Gut Health Team (LiGHT) Ghent, Ghent University, 9820, Merelbeke, Belgium.
| |
Collapse
|
3
|
Várnagy Á, Mauchart P, Nagy G, Bódis J, Sulyok E. Follicular Fluid Vanin-1 Levels in Patients Undergoing Ivf: A Preliminary Study. Antioxidants (Basel) 2025; 14:133. [PMID: 40002320 PMCID: PMC11851465 DOI: 10.3390/antiox14020133] [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: 11/29/2024] [Revised: 12/27/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
This preliminary study was designed to determine follicular fluid (FF) vanin-1 levels, to assess their relation to serum vanin-1 and to reveal their potential to predict the outcome of in vitro fertilization (IVF). Eighteen unselected, consecutive women undergoing IVF were included. Serum and pooled FF samples were obtained simultaneously during routine IVF procedures. Vanin-1 levels were measured by using commercially available ELISA kits. As most of the values were below 0.6 ng/mL, the data are given as optical density. It was found that vanin-1 can be detected in FF and that it is not significantly related to its maternal serum levels (p = 0.06). FF vanin-1 levels proved to be higher in non-pregnant as compared to pregnant women (p < 0.04). There are significant positive relationships between the FF to serum vanin-1 ratio and body mass index (BMI, p < 0.02), anti-Müllerian hormone (AMH, p < 0.02) and baseline serum estradiol (p < 0.01). Moreover, the FF/serum vanin-1 ratio tended to increase with cumulative FSH dose, but this increase did not reach statistical significance (p = 0.064). It may be concluded that FF vanin-1 may serve as a biomarker to predict IVF outcome. To confirm this contention, further studies are to be performed.
Collapse
Affiliation(s)
- Ákos Várnagy
- National Laboratory on Human Reproduction, University of Pécs, H-7624 Pécs, Hungary; (Á.V.); (J.B.); (E.S.)
- HUN-REN–PTE Human Reproduction Research Group, H-7624 Pécs, Hungary
- Department of Obstetrics and Gynecology, School of Medicine, University of Pécs, H-7624 Pécs, Hungary
| | - Péter Mauchart
- National Laboratory on Human Reproduction, University of Pécs, H-7624 Pécs, Hungary; (Á.V.); (J.B.); (E.S.)
- HUN-REN–PTE Human Reproduction Research Group, H-7624 Pécs, Hungary
- Department of Obstetrics and Gynecology, School of Medicine, University of Pécs, H-7624 Pécs, Hungary
| | - Gábor Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - József Bódis
- National Laboratory on Human Reproduction, University of Pécs, H-7624 Pécs, Hungary; (Á.V.); (J.B.); (E.S.)
- HUN-REN–PTE Human Reproduction Research Group, H-7624 Pécs, Hungary
- Department of Obstetrics and Gynecology, School of Medicine, University of Pécs, H-7624 Pécs, Hungary
- Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, H-7624 Pécs, Hungary
| | - Endre Sulyok
- National Laboratory on Human Reproduction, University of Pécs, H-7624 Pécs, Hungary; (Á.V.); (J.B.); (E.S.)
- HUN-REN–PTE Human Reproduction Research Group, H-7624 Pécs, Hungary
- Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
4
|
Dai D, Zhang Z, Ma M, Li J, Zhang S, Ma P, Song D. Vanin-1-Activated Fluorescent Probe for Real-Time In Vivo Imaging of Inflammatory Responses Across Multiple Tissue Types. Anal Chem 2025; 97:1402-1409. [PMID: 39787461 DOI: 10.1021/acs.analchem.4c05982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Vanin-1 is a pantetheine hydrolase that plays a key role in inflammatory diseases. Effective tools for noninvasive, real-time monitoring of Vanin-1 are lacking, largely due to background fluorescence interference in existing probes. To address this issue, we developed a dual-modal fluorescent and colorimetric probe, MB-Van1, to detect Vanin-1 with high sensitivity and selectivity. MB-Van1 has a structure optimized to exhibit nearly zero background fluorescence, resulting in a high signal-to-noise ratio that enables the accurate detection of Vanin-1 activity in various biological tissues. In vitro experiments demonstrated that MB-Van1 had a detection limit as low as 0.031 ng/mL in the fluorescence mode. We successfully employ MB-Van1 to observe elevated Vanin-1 levels in inflammatory tissues of various mouse models of rheumatoid arthritis (RA), drug-induced liver injury (DILI), and nonsteroidal anti-inflammatory drug (NSAID) enteropathy models, within only 5 min. This advancement provides a novel approach for monitoring the dynamic changes of Vanin-1 during inflammation, offering new strategies for the early diagnosis and therapeutic assessment of other related diseases.
Collapse
Affiliation(s)
- Dianfeng Dai
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Zhimin Zhang
- Department of Pharmacy, Changchun Medical College, Changchun 130031, China
| | - Mo Ma
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
- School of Pharmacy, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Jingkang Li
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Siqi Zhang
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Pinyi Ma
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| | - Daqian Song
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Qianjin Street 2699, Changchun 130012, China
| |
Collapse
|
5
|
Xie T, Cao GY, Zhang S, Li MK, Jin X, Liu L, Wang G, Zhen L. Discovery of Thiazole Carboxamides as Novel Vanin-1 Inhibitors for Inflammatory Bowel Disease Treatment. J Med Chem 2024; 67:20372-20398. [PMID: 39514323 DOI: 10.1021/acs.jmedchem.4c01838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Inflammatory bowel disease (IBD) is a clinically heterogeneous disease demanding more therapeutic targets and intervention strategies. Vanin-1, an oxidative stress-regulating protein, has emerged as a promising target for alleviating inflammation and oxidative stress. In this study, a series of thiazole carboxamide derivatives as vanin-1 inhibitors were designed and synthesized. The preferred compound, X17, demonstrated potent inhibition against vanin-1 at the protein, HT-29 cell, and tissue levels, whose binding mode with the target was confirmed via the cocrystal structure. X17 achieved a high bioavailability of 81% in rats, accompanied by concentration-dependent inhibition of serum vanin-1. In a DSS-induced mouse colitis model, X17 exhibited potent anti-inflammatory and antioxidant activities, repressing the inflammatory factor expressions and myeloperoxidase activity, elevating the colonic glutathione reserve, and restoring the intestinal barrier. Collectively, these findings depict the discovery of a potent vanin-1 inhibitor, providing an opportunity for further drug candidate development for treating IBD.
Collapse
Affiliation(s)
- Tao Xie
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Gao-Yao Cao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Shize Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Meng-Ke Li
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Xin Jin
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Liu Liu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Le Zhen
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
6
|
Kumaragurubaran N, Huang YZ, Mockaitis T, Arul P, Huang ST, Lin HY, Wei YC, Morkvenaite-Vilkonciene I. Development of an Activity-Based Ratiometric Electrochemical Switch for Direct, Real-Time Sensing of Pantetheinase in Live Cells, Blood, and Urine Samples. ACS Sens 2024; 9:5436-5444. [PMID: 39331818 PMCID: PMC11519916 DOI: 10.1021/acssensors.4c01658] [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: 07/04/2024] [Revised: 08/30/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024]
Abstract
Pantetheinase is a key biomarker for the diagnosis of acute kidney injury and the monitoring of malaria progression. Currently, existing methods for sensing pantetheinase, also known as Vanin-1, show considerable potential but come with certain limitations, including their inability to directly sense analytes in turbid biofluid samples without tedious sample pretreatment. Here, we describe the first activity-based electrochemical probe, termed VaninLP, for convenient and specific direct targeting of pantetheinase activity in turbid liquid biopsy samples. The probe was designed such that cleavage of the pantetheinase amide linkage, triggered by a self-immolative reaction, simultaneously ejects an amino ferrocene reporter. Among the distinctive properties of the VaninLP probe for sensing pantetheinase are its high selectivity, sensitivity, and enzyme affinity, a wide linear concentration range (8-300 ng/mL), and low limit of detection (2.47 ng/mL). The designed probe precisely targeted pantetheinase and was free of interference by other electroactive biological species. We further successfully applied the VaninLP probe to monitor and quantify the activity of pantetheinase on the surfaces of HepG2 tumor cells, blood, and urine samples. Collectively, our findings indicate that VaninLP holds significant promise as a point-of-care tool for diagnosing early-stage kidney injury, as well as monitoring the progression of malaria.
Collapse
Affiliation(s)
- Namasivayam Kumaragurubaran
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 106, Taiwan, ROC
- Institute
of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei 106, Taiwan, ROC
| | - Yan-Zhi Huang
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 106, Taiwan, ROC
- Institute
of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei 106, Taiwan, ROC
| | - Tomas Mockaitis
- Department
of Nanotechnology, State Research Institute
Centre for Physical Sciences and Technology (FTMC), Sauletekio av. 3, Vilnius 10257, Lithuania
| | - Ponnusamy Arul
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 106, Taiwan, ROC
- Institute
of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei 106, Taiwan, ROC
| | - Sheng-Tung Huang
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 106, Taiwan, ROC
- Institute
of Biochemical and Biomedical Engineering, National Taipei University of Technology, Taipei 106, Taiwan, ROC
- High-Value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, No. 1, Sec. 3, Zhongxiao E. Rd., Taipei 10608, Taiwan, ROC
| | - Hsin-Yi Lin
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 106, Taiwan, ROC
| | - Yi-Cheng Wei
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 106, Taiwan, ROC
| | - Inga Morkvenaite-Vilkonciene
- Department
of Nanotechnology, State Research Institute
Centre for Physical Sciences and Technology (FTMC), Sauletekio av. 3, Vilnius 10257, Lithuania
| |
Collapse
|
7
|
Munteanu C, Mârza SM, Papuc I. The immunomodulatory effects of vitamins in cancer. Front Immunol 2024; 15:1464329. [PMID: 39434876 PMCID: PMC11491384 DOI: 10.3389/fimmu.2024.1464329] [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: 07/13/2024] [Accepted: 09/17/2024] [Indexed: 10/23/2024] Open
Abstract
Nutrition may affect animal health due to the strong link between them. Also, diets improve the healing process in various disease states. Cancer is a disease, where the harmful consequences of tumors severely impair the body. The information regarding the evolution of this disease is extrapolated from human to animal because there are few specific studies regarding nutritional needs in animals with cancer. Thus, this paper aims to review the literature regarding the immunomodulatory effects of vitamins in mammal cancer. An adequate understanding of the metabolism and requirements of nutrients for mammals is essential to ensuring their optimal growth, development, and health, regardless of their food sources. According to these: 1) Some species are highly dependent on vitamin D from food, so special attention must be paid to this aspect. Calcitriol/VDR signaling can activate pro-apoptotic proteins and suppress anti-apoptotic ones. 2) Nitric oxide (NO) production is modulated by vitamin E through inhibiting transcription nuclear factor kappa B (NF-κB) activation. 3) Thiamine supplementation could be responsible for the stimulation of tumor cell proliferation, survival, and resistance to chemotherapy. 4) Also, it was found that the treatment with NO-Cbl in dogs is a viable anti-cancer therapy that capitalizes on the tumor-specific properties of the vitamin B12 receptor. Therefore, diets should contain the appropriate class of compounds in adequate proportions. Also, the limitations of this paper are that some vitamins are intensively studied and at the same time regarding others, there is a lack of information, especially in animals. Therefore, some subsections are longer and more heavily debated than others.
Collapse
Affiliation(s)
- Camelia Munteanu
- Department of Plant Culture, Faculty of Agriculture, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, ;Romania
| | - Sorin Marian Mârza
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, Cluj-Napoca, ;Romania
| | - Ionel Papuc
- Faculty of Veterinary Medicine, University of Agricultural Science and Veterinary Medicine, Cluj-Napoca, ;Romania
| |
Collapse
|
8
|
Tserga A, Saulnier-Blache JS, Palamaris K, Pouloudi D, Gakiopoulou H, Zoidakis J, Schanstra JP, Vlahou A, Makridakis M. Complement Cascade Proteins Correlate with Fibrosis and Inflammation in Early-Stage Type 1 Diabetic Kidney Disease in the Ins2Akita Mouse Model. Int J Mol Sci 2024; 25:1387. [PMID: 38338666 PMCID: PMC10855735 DOI: 10.3390/ijms25031387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
Diabetic kidney disease (DKD) is characterized by histological changes including fibrosis and inflammation. Evidence supports that DKD is mediated by the innate immune system and more specifically by the complement system. Using Ins2Akita T1D diabetic mice, we studied the connection between the complement cascade, inflammation, and fibrosis in early DKD. Data were extracted from a previously published quantitative-mass-spectrometry-based proteomics analysis of kidney glomeruli of 2 (early DKD) and 4 months (moderately advanced DKD)-old Ins2Akita mice and their controls A Spearman rho correlation analysis of complement- versus inflammation- and fibrosis-related protein expression was performed. A cross-omics validation of the correlation analyses' results was performed using public-domain transcriptomics datasets (Nephroseq). Tissue sections from 43 patients with DKD were analyzed using immunofluorescence. Among the differentially expressed proteins, the complement cascade proteins C3, C4B, and IGHM were significantly increased in both early and later stages of DKD. Inflammation-related proteins were mainly upregulated in early DKD, and fibrotic proteins were induced in moderately advanced stages of DKD. The abundance of complement proteins with fibrosis- and inflammation-related proteins was mostly positively correlated in early stages of DKD. This was confirmed in seven additional human and mouse transcriptomics DKD datasets. Moreover, C3 and IGHM mRNA levels were found to be negatively correlated with the estimated glomerular filtration rate (range for C3 rs = -0.58 to -0.842 and range for IGHM rs = -0.6 to -0.74) in these datasets. Immunohistology of human kidney biopsies revealed that C3, C1q, and IGM proteins were induced in patients with DKD and were correlated with fibrosis and inflammation. Our study shows for the first time the potential activation of the complement cascade associated with inflammation-mediated kidney fibrosis in the Ins2Akita T1D mouse model. Our findings could provide new perspectives for the treatment of early DKD as well as support the use of Ins2Akita T1D in pre-clinical studies.
Collapse
Affiliation(s)
- Aggeliki Tserga
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (J.Z.); (A.V.)
| | - Jean Sébastien Saulnier-Blache
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France; (J.S.S.-B.); (J.P.S.)
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Kostantinos Palamaris
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 34400 Athens, Greece; (K.P.); (D.P.); (H.G.)
| | - Despoina Pouloudi
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 34400 Athens, Greece; (K.P.); (D.P.); (H.G.)
| | - Harikleia Gakiopoulou
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 34400 Athens, Greece; (K.P.); (D.P.); (H.G.)
| | - Jerome Zoidakis
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (J.Z.); (A.V.)
- Department of Biology, National and Kapodistrian University of Athens, 15701 Zografou, Greece
| | - Joost Peter Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France; (J.S.S.-B.); (J.P.S.)
- Department of Biology, Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Antonia Vlahou
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (J.Z.); (A.V.)
| | - Manousos Makridakis
- Biomedical Research Foundation, Academy of Athens, Department of Biotechnology, Soranou Efessiou 4, 11527 Athens, Greece; (A.T.); (J.Z.); (A.V.)
| |
Collapse
|
9
|
Qi P, Huang M, Ren X, Zhai Y, Qiu C, Zhu H. Identification of potential biomarkers and therapeutic targets related to post-traumatic stress disorder due to traumatic brain injury. Eur J Med Res 2024; 29:44. [PMID: 38212778 PMCID: PMC10782540 DOI: 10.1186/s40001-024-01640-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Post-traumatic stress disorder (PTSD), a disease state that has an unclear pathogenesis, imposes a substantial burden on individuals and society. Traumatic brain injury (TBI) is one of the most significant triggers of PTSD. Identifying biomarkers associated with TBI-related PTSD will help researchers to uncover the underlying mechanism that drives disease development. Furthermore, it remains to be confirmed whether different types of traumas share a common mechanism of action. METHODS For this study, we screened the eligible data sets from the Gene Expression Omnibus (GEO) database, obtained differentially expressed genes (DEGs) through analysis, conducted functional enrichment analysis on the DEGs in order to understand their molecular mechanisms, constructed a PPI network, used various algorithms to obtain hub genes, and finally evaluated, validated, and analyzed the diagnostic performance of the hub genes. RESULTS A total of 430 upregulated and 992 down-regulated differentially expressed genes were extracted from the TBI data set. A total of 1919 upregulated and 851 down-regulated differentially expressed genes were extracted from the PTSD data set. Functional enrichment analysis revealed that the differentially expressed genes had biological functions linked to molecular regulation, cell signaling transduction, cell metabolic regulation, and immune response. After constructing a PPI network and introducing algorithm analysis, the upregulated hub genes were identified as VNN1, SERPINB2, and ETFDH, and the down-regulated hub genes were identified as FLT3LG, DYRK1A, DCN, and FKBP8. In addition, by comparing the data with patients with other types of trauma, it was revealed that PTSD showed different molecular processes that are under the influence of different trauma characteristics and responses. CONCLUSIONS By exploring the role of different types of traumas during the pathogenesis of PTSD, its possible molecular mechanisms have been revealed, providing vital information for understanding the complex pathways associated with TBI-related PTSD. The data in this study has important implications for the design and development of new diagnostic and therapeutic methods needed to treat and manage PTSD.
Collapse
Affiliation(s)
- Peng Qi
- Department of Emergency, First Medical Center of Chinese, PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Mengjie Huang
- Department of Nephrology, First Medical Center of Chinese, PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Xuewen Ren
- Department of Emergency, First Medical Center of Chinese, PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Yongzhi Zhai
- Department of Emergency, First Medical Center of Chinese, PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Chen Qiu
- Department of Orthopedics, Fourth Medical Center of Chinese, PLA General Hospital, Beijing, 100853, China.
| | - Haiyan Zhu
- Department of Emergency, First Medical Center of Chinese, PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
10
|
Yu H, Cui Y, Guo F, Zhu Y, Zhang X, Shang D, Dong D, Xiang H. Vanin1 (VNN1) in chronic diseases: Future directions for targeted therapy. Eur J Pharmacol 2024; 962:176220. [PMID: 38042463 DOI: 10.1016/j.ejphar.2023.176220] [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: 07/15/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 12/04/2023]
Abstract
Vanin1 (VNN1) is an exogenous enzyme with pantetheinase activity that mainly exerts physiological functions through enzyme catalysis products, including pantothenic acid and cysteamine. In recent years, the crosstalk between VNN1 and metabolism and oxidative stress has attracted much attention. As a result of the ability of VNN1 to affect multiple metabolic pathways and oxidative stress to exacerbate or alleviate pathological processes, it has become a key component of disease progression. This review discusses the functions of VNN1 in glucolipid metabolism, cysteamine metabolism, and glutathione metabolism to provide perspectives on VNN1-targeted therapy for chronic diseases.
Collapse
Affiliation(s)
- Hao Yu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, 116011, China; College of Pharmacy, Dalian Medical University, 116044, China
| | - Yuying Cui
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Fangyue Guo
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - YuTong Zhu
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Xiaonan Zhang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Dong Shang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China; Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, 116011, China.
| | - Hong Xiang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
11
|
Zamora Z, Wang S, Chen YW, Diamante G, Yang X. Systematic transcriptome-wide meta-analysis across endocrine disrupting chemicals reveals shared and unique liver pathways, gene networks, and disease associations. ENVIRONMENT INTERNATIONAL 2024; 183:108339. [PMID: 38043319 PMCID: PMC11216742 DOI: 10.1016/j.envint.2023.108339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/03/2023] [Accepted: 11/19/2023] [Indexed: 12/05/2023]
Abstract
Cardiometabolic disorders (CMD) are a growing public health problem across the world. Among the known cardiometabolic risk factors are compounds that induce endocrine and metabolic dysfunctions, such as endocrine disrupting chemicals (EDCs). To date, how EDCs influence molecular programs and cardiometabolic risks has yet to be fully elucidated, especially considering the complexity contributed by species-, chemical-, and dose-specific effects. Moreover, different experimental and analytical methodologies employed by different studies pose challenges when comparing findings across studies. To explore the molecular mechanisms of EDCs in a systematic manner, we established a data-driven computational approach to meta-analyze 30 human, mouse, and rat liver transcriptomic datasets for 4 EDCs, namely bisphenol A (BPA), bis(2-ethylhexyl) phthalate (DEHP), tributyltin (TBT), and perfluorooctanoic acid (PFOA). Our computational pipeline uniformly re-analyzed pre-processed quality-controlled microarray data and raw RNAseq data, derived differentially expressed genes (DEGs) and biological pathways, modeled gene regulatory networks and regulators, and determined CMD associations based on gene overlap analysis. Our approach revealed that DEHP and PFOA shared stable transcriptomic signatures that are enriched for genes associated with CMDs, suggesting similar mechanisms of action such as perturbations of peroxisome proliferator-activated receptor gamma (PPARγ) signaling and liver gene network regulators VNN1 and ACOT2. In contrast, TBT exhibited highly divergent gene signatures, pathways, network regulators, and disease associations from the other EDCs. In addition, we found that the rat, mouse, and human BPA studies showed highly variable transcriptomic patterns, providing molecular support for the variability in BPA responses. Our work offers insights into the commonality and differences in the molecular mechanisms of various EDCs and establishes a streamlined data-driven workflow to compare molecular mechanisms of environmental substances to elucidate the underlying connections between chemical exposure and disease risks.
Collapse
Affiliation(s)
- Zacary Zamora
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Susanna Wang
- Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Yen-Wei Chen
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| | - Xia Yang
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Integrative Biology and Physiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA.
| |
Collapse
|
12
|
Kishimoto Y, Saita E, Ohmori R, Kondo K, Momiyama Y. High plasma concentrations of vanin-1 in patients with coronary artery disease. Heart Vessels 2024; 39:10-17. [PMID: 37582951 DOI: 10.1007/s00380-023-02305-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/09/2023] [Indexed: 08/17/2023]
Abstract
Vanin-1 is a pantetheinase that hydrolyzes pantetheine to pantothenic acid and cysteamine. Vanin-1 has become recognized to be associated with oxidative stress and inflammation. In animal models, vanin-1 was reported to accelerate atherosclerosis. However, no study has reported blood vanin-1 concentrations in patients with coronary artery disease (CAD). We investigated plasma vanin-1 concentrations in 388 patients undergoing elective coronary angiography for suspected CAD. Patients with acute coronary syndrome were excluded. Of the 388 study patients, CAD was found in 207 patients [1-vessel (1-VD), n = 88; 2-vessel (2-VD), n = 66; and 3-vessel disease (3-VD), n = 53]. Plasma vanin-1 concentrations were higher in patients with CAD than in those without CAD (median 0.59 vs. 0.46 ng/mL, P < 0.005). Vanin-1 concentrations in patients without CAD and those with 1-VD, 2-VD, and 3-VD were 0.46, 0.58, 0.57, and 0.61 ng/mL, respectively, and were highest in 3-VD (P < 0.05). A high vainin-1 concentration (> 0.48 ng/mL) was found in 46% of patients without CAD, 61% of 1-VD, 65% of 2-VD, and 66% of 3-VD (P < 0.01). Vanin-1 concentrations significantly correlated with the number of stenotic coronary segments (r = 0.14, P < 0.02). In the multivariate analysis, vanin-1 concentration was a significant factor associated with CAD independent of atherosclerotic risk factors. The odds ratio for CAD was 1.63 (95%CI = 1.04-2.55) for the high vanin-1 concentration of > 0.48 ng/mL. Thus, plasma vanin-1 concentrations in patients with CAD were found to be high and to be associated with the presence and severity of CAD.
Collapse
Affiliation(s)
- Yoshimi Kishimoto
- Department of Food Science and Human Nutrition, Setsunan University, Osaka, Japan
| | - Emi Saita
- Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan
| | - Reiko Ohmori
- Faculty of Regional Design, Utsunomiya University, Tochigi, Japan
| | | | - Yukihiko Momiyama
- Department of Cardiology, National Hospital Organization Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo, 152-8902, Japan.
| |
Collapse
|
13
|
Rezende TMT, Menezes HSG, Rezende AM, Cavalcanti MP, Silva YMG, de-Melo-Neto OP, Romão TP, Silva-Filha MHNL. Culex quinquefasciatus Resistant to the Binary Toxin from Lysinibacillus sphaericus Displays a Consistent Downregulation of Pantetheinase Transcripts. Biomolecules 2023; 14:33. [PMID: 38254633 PMCID: PMC10813629 DOI: 10.3390/biom14010033] [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: 09/29/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Culex quinquefasciatus resistance to the binary (Bin) toxin, the major larvicidal component from Lysinibacillus sphaericus, is associated with mutations in the cqm1 gene, encoding the Bin-toxin receptor. Downregulation of the cqm1 transcript was found in the transcriptome of larvae resistant to the L. sphaericus IAB59 strain, which produces both the Bin toxin and a second binary toxin, Cry48Aa/Cry49Aa. Here, we investigated the transcription profiles of two other mosquito colonies having Bin resistance only. These confirmed the cqm1 downregulation and identified transcripts encoding the enzyme pantetheinase as the most downregulated mRNAs in both resistant colonies. Further quantification of these transcripts reinforced their strong downregulation in Bin-resistant larvae. Multiple genes were found encoding this enzyme in Cx. quinquefasciatus and a recombinant pantetheinase was then expressed in Escherichia coli and Sf9 cells, with its presence assessed in the midgut brush border membrane of susceptible larvae. The pantetheinase was expressed as a ~70 kDa protein, potentially membrane-bound, which does not seem to be significantly targeted by glycosylation. This is the first pantetheinase characterization in mosquitoes, and its remarkable downregulation might reflect features impacted by co-selection with the Bin-resistant phenotype or potential roles in the Bin-toxin mode of action that deserve to be investigated.
Collapse
Affiliation(s)
- Tatiana M. T. Rezende
- Department of Entomology, Instituto Aggeu Magalhães-Fiocruz, Recife 50740-465, PE, Brazil; (T.M.T.R.); (H.S.G.M.); (Y.M.G.S.); (T.P.R.)
| | - Heverly S. G. Menezes
- Department of Entomology, Instituto Aggeu Magalhães-Fiocruz, Recife 50740-465, PE, Brazil; (T.M.T.R.); (H.S.G.M.); (Y.M.G.S.); (T.P.R.)
| | - Antonio M. Rezende
- Department of Microbiology, Instituto Aggeu Magalhães-Fiocruz, Recife 50740-465, PE, Brazil; (A.M.R.); (M.P.C.); (O.P.d.-M.-N.)
| | - Milena P. Cavalcanti
- Department of Microbiology, Instituto Aggeu Magalhães-Fiocruz, Recife 50740-465, PE, Brazil; (A.M.R.); (M.P.C.); (O.P.d.-M.-N.)
| | - Yuri M. G. Silva
- Department of Entomology, Instituto Aggeu Magalhães-Fiocruz, Recife 50740-465, PE, Brazil; (T.M.T.R.); (H.S.G.M.); (Y.M.G.S.); (T.P.R.)
| | - Osvaldo P. de-Melo-Neto
- Department of Microbiology, Instituto Aggeu Magalhães-Fiocruz, Recife 50740-465, PE, Brazil; (A.M.R.); (M.P.C.); (O.P.d.-M.-N.)
| | - Tatiany P. Romão
- Department of Entomology, Instituto Aggeu Magalhães-Fiocruz, Recife 50740-465, PE, Brazil; (T.M.T.R.); (H.S.G.M.); (Y.M.G.S.); (T.P.R.)
| | - Maria Helena N. L. Silva-Filha
- Department of Entomology, Instituto Aggeu Magalhães-Fiocruz, Recife 50740-465, PE, Brazil; (T.M.T.R.); (H.S.G.M.); (Y.M.G.S.); (T.P.R.)
- National Institute for Molecular Entomology, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
14
|
Feng Y, Xu S, Guo H, Ren TB, Huan SY, Yuan L, Zhang XB. Vanin-1-Activated Chemiluminescent Probe: Help to Early Diagnosis of Acute Kidney Injury with High Signal-to-Noise Ratio through Urinalysis. Anal Chem 2023; 95:14754-14761. [PMID: 37734030 DOI: 10.1021/acs.analchem.3c02875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
Acute kidney injury (AKI) is a common medical condition with high morbidity and mortality. Although urinalysis provides a noninvasive and convenient diagnostic method for AKI at the molecular level, the low sensitivity of current chemical probes used in urinalysis hinders the time diagnosis of AKI. Herein, we achieved the sensitive and early diagnosis of AKI by the development of a chemiluminescent probe CL-Pa suitable for detection of urinary Vanin-1. Vanin-1 is considered as an early and sensitive biomarker for AKI, while few chemical probes have been applied to for its efficient detection. By virtue of the low autofluorescence interference during urine imaging in the chemiluminescence model, CL-Pa could realize the monitoring of the up-regulated urinary Vanin-1 with a high signal-to-noise ratio (∼588). Importantly, under the help of CL-Pa, the up-regulation of urinary Vanin-1 of cisplatin-induced AKI mice at 12 h post cisplatin injection was detected, which was much earlier than clinical biomarkers (sCr and BUN) and change of kidney histology (48 h post cisplatin injection). Furthermore, using this probe, the fluctuation of urinary Vanin-1 of mice with different degrees of AKI was monitored. This study demonstrated the ability of CL-Pa in sensitively detecting drug-induced AKI through urinalysis and suggested the great potential of CL-Pa for early diagnosis of AKI and evaluate the efficiency of anti-AKI drugs clinically.
Collapse
Affiliation(s)
- Yurong Feng
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, P. R. China
| | - Shuai Xu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, P. R. China
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Haowei Guo
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, P. R. China
| | - Tian-Bing Ren
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, P. R. China
| | - Shuang-Yan Huan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, P. R. China
| | - Lin Yuan
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, P. R. China
| | - Xiao-Bing Zhang
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Collaborative Innovation Center for Chemistry and Molecular Medicine, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
15
|
Miallot R, Millet V, Galland F, Naquet P. The vitamin B5/coenzyme A axis: A target for immunomodulation? Eur J Immunol 2023; 53:e2350435. [PMID: 37482959 DOI: 10.1002/eji.202350435] [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: 04/05/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
Coenzyme A (CoA) serves as a vital cofactor in numerous enzymatic reactions involved in energy production, lipid metabolism, and synthesis of essential molecules. Dysregulation of CoA-dependent metabolic pathways can contribute to chronic diseases, such as inflammatory diseases, obesity, diabetes, cancer, and cardiovascular disorders. Additionally, CoA influences immune cell activation by modulating the metabolism of these cells, thereby affecting their proliferation, differentiation, and effector functions. Targeting CoA metabolism presents a promising avenue for therapeutic intervention, as it can potentially restore metabolic balance, mitigate chronic inflammation, and enhance immune cell function. This might ultimately improve the management and outcomes for these diseases. This review will more specifically focus on the contribution of pathways regulating the availability of the CoA precursor Vitamin B5/pantothenate in vivo and modulating the development of Th17-mediated inflammation, CD8-dependent anti-tumor immunity but also tissue repair processes in chronic inflammatory or degenerative diseases.
Collapse
|
16
|
Patel B, Eskander MA, Fang-Mei Chang P, Chapa B, Ruparel SB, Lai Z, Chen Y, Akopian A, Ruparel NB. Understanding painful versus non-painful dental pain in female and male patients: A transcriptomic analysis of human biopsies. PLoS One 2023; 18:e0291724. [PMID: 37733728 PMCID: PMC10513205 DOI: 10.1371/journal.pone.0291724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
Dental pain from apical periodontitis is an infection induced-orofacial pain condition that presents with diversity in pain phenotypes among patients. While 60% of patients with a full-blown disease present with the hallmark symptom of mechanical allodynia, nearly 40% of patients experience no pain. Furthermore, a sexual dichotomy exists, with females exhibiting lower mechanical thresholds under basal and diseased states. Finally, the prevalence of post-treatment pain refractory to commonly used analgesics ranges from 7-19% (∼2 million patients), which warrants a thorough investigation of the cellular changes occurring in different patient cohorts. We, therefore, conducted a transcriptomic assessment of periapical biopsies (peripheral diseased tissue) from patients with persistent apical periodontitis. Surgical biopsies from symptomatic male (SM), asymptomatic male (AM), symptomatic female (SF), and asymptomatic female (AF) patients were collected and processed for bulk RNA sequencing. Using strict selection criteria, our study found several unique differentially regulated genes (DEGs) between symptomatic and asymptomatic patients, as well as novel candidate genes between sexes within the same pain group. Specifically, we found the role of cells of the innate and adaptive immune system in mediating nociception in symptomatic patients and the role of genes involved in tissue homeostasis in potentially inhibiting nociception in asymptomatic patients. Furthermore, sex-related differences appear to be tightly regulated by macrophage activity, its secretome, and/or migration. Collectively, we present, for the first time, a comprehensive assessment of peripherally diseased human tissue after a microbial insult and shed important insights into the regulation of the trigeminal system in female and male patients.
Collapse
Affiliation(s)
- Biraj Patel
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Michael A. Eskander
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Phoebe Fang-Mei Chang
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Brett Chapa
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Shivani B. Ruparel
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Zhao Lai
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Armen Akopian
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Nikita B. Ruparel
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
17
|
Zhao D, Qin D, Yin L, Yang Q. Integrated Bioinformatics Analysis and Experimental Verification of Immune Cell Infiltration and the Related Core Genes in Ulcerative Colitis. Pharmgenomics Pers Med 2023; 16:629-643. [PMID: 37383675 PMCID: PMC10296601 DOI: 10.2147/pgpm.s406644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/26/2023] [Indexed: 06/30/2023] Open
Abstract
Background Ulcerative colitis is a recurrent autoimmune disease. At present, the pathogenesis of UC is not completely clear. Hence, the etiology and underlying molecular mechanism need to be further investigated. Methods Three sets of microarray datasets were included from the Gene Expression Omnibus database. The differentially expressed genes in two sets of datasets were analyzed using the R software, and the core genes of UC were screened using machine learning. The sensitivity and specificity of the core genes were evaluated with the receiver operating characteristic curve in another microarray dataset. Subsequently, the CIBERSORT tool was used to analyze the relationship between UC and its core genes and immune cell infiltration. To verify the relationship between UC and core genes and the relationship between core genes and immune cell infiltration in vivo. Results A total of 36 DEGs were identified. AQP8, HMGCS2, and VNN1 were determined to be the core genes of UC. These genes had high sensitivity and specificity in receiver operating characteristic curve analysis. According to the analysis of immune cell infiltration, neutrophils, monocytes, and macrophages were positively correlated with UC. AQP8, HMGCS2, and VNN1 were also correlated with immune cell infiltration to varying degrees. In vivo experiments verified that the expressions of neutrophils, monocytes, and macrophages increased in the UC colon. Furthermore, the expressions of AQP8 and HMGCS2 decreased, whereas that of VNN1 increased. Azathioprine treatment improved all the indicators to different degrees. Conclusion AQP8, HMGCS2, and VNN1 are the core genes of UC and exhibit different degrees of correlation with immune cells. These genes are expected to become new therapeutic targets for UC. Moreover, the occurrence and development of UC are influenced by immune cell infiltration.
Collapse
Affiliation(s)
- Danya Zhao
- The First School of Clinical Medicine of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Danping Qin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Liming Yin
- Institute of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Qiang Yang
- Department of Gastroenterology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| |
Collapse
|
18
|
Chen C, Zuo Y, Hu H, Li X, Zhang L, Yang D, Liu F, Liao X, Xiong G, Cao Z, Zhong Z, Bi Y, Lu H, Chen J. Hepatic lipid metabolism disorders and immunotoxicity induced by cysteamine in early developmental stages of zebrafish. Toxicology 2023; 493:153555. [PMID: 37236339 DOI: 10.1016/j.tox.2023.153555] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/13/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023]
Abstract
Cysteamine, a sulfhydryl compound, is an intermediate in the metabolism of coenzyme A to taurine in living organisms. However, the potential side effects of cysteamine such as hepatotoxicity in pediatric patients have been reported in some studies. To evaluate the impact of cysteamine on infants and children, larval zebrafish (a vertebrate model) were exposed to 0.18, 0.36 and 0.54mM cysteamine from 72 hpf to 144 hpf. Alterations in general and pathological evaluation, biochemical parameters, cell proliferation, lipid metabolism factors, inflammatory factors and Wnt signaling pathway levels were examined. Increased liver area and lipid accumulation were observed in liver morphology, staining and histopathology in a dose-dependent manner with cysteamine exposure. In addition, the experimental cysteamine group exhibited higher alanine aminotransferase, aspartate aminotransferase, total triglyceride and total cholesterol levels than the control group. Meanwhile, the levels of lipogenesis-related factors ascended whereas lipid transport-related factors descended. Oxidative stress indicators such as reactive oxygen species, MDA and SOD were upregulated after cysteamine exposure. Afterwards, transcription assays revealed that biotinidase and Wnt pathway-related genes were upregulated in the exposed group, and inhibition of Wnt signaling partially rescued the abnormal liver development. The current study found that cysteamine-induced hepatotoxicity in larval zebrafish is due to inflammation and abnormal lipid metabolism, which is mediated by biotinidase (a potential pantetheinase isoenzyme) and Wnt signaling. This provides a perspective on the safety of cysteamine administration in children and identifies potential targets for protection against adverse reactions.
Collapse
Affiliation(s)
- Chao Chen
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai, 200092, China; Department of Ophthalmology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yuhua Zuo
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai, 200092, China
| | - Hongmei Hu
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai, 200092, China
| | - Xue Li
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai, 200092, China
| | - Li Zhang
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai, 200092, China
| | - Dou Yang
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China
| | - Fasheng Liu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China
| | - Xinjun Liao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China
| | - Guanghua Xiong
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China
| | - Zigang Cao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China
| | - Zilin Zhong
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yanlong Bi
- Department of Ophthalmology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China.
| | - Huiqiang Lu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, Clinical Research Center of Affiliated Hospital of Jinggangshan University, College of Life Sciences, Jinggangshan University, Ji'an, 343009, Jiangxi, China.
| | - Jianjun Chen
- Translational Research Institute of Brain and Brain-like Intelligence, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Department of Pediatrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
19
|
Küçük ŞD, Gezer E, Çalan M, Yüksel A. Relationship between serum Vanin 1 level and metabolic parameters in patients with type 2 diabetes mellitus. Int J Diabetes Dev Ctries 2022. [DOI: 10.1007/s13410-022-01153-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
20
|
Millet V, Gensollen T, Maltese M, Serrero M, Lesavre N, Bourges C, Pitaval C, Cadra S, Chasson L, Vu Man TP, Masse M, Martinez-Garcia JJ, Tranchida F, Shintu L, Mostert K, Strauss E, Lepage P, Chamaillard M, Broggi A, Peyrin-Biroulet L, Grimaud JC, Naquet P, Galland F. Harnessing the Vnn1 pantetheinase pathway boosts short chain fatty acids production and mucosal protection in colitis. Gut 2022; 72:1115-1128. [PMID: 36175116 DOI: 10.1136/gutjnl-2021-325792] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/05/2022] [Indexed: 12/26/2022]
Abstract
OBJECTIVE In the management of patients with IBD, there is a need to identify prognostic markers and druggable biological pathways to improve mucosal repair and probe the efficacy of tumour necrosis factor alpha biologics. Vnn1 is a pantetheinase that degrades pantetheine to pantothenate (vitamin B5, a precursor of coenzyme A (CoA) biosynthesis) and cysteamine. Vnn1 is overexpressed by inflamed colonocytes. We investigated its contribution to the tolerance of the intestinal mucosa to colitis-induced injury. DESIGN We performed an RNA sequencing study on colon biopsy samples from patients with IBD stratified according to clinical severity and modalities of treatment. We generated the VIVA mouse transgenic model, which specifically overexpresses Vnn1 on intestinal epithelial cells and explored its susceptibility to colitis. We developed a pharmacological mimicry of Vnn1 overexpression by administration of Vnn1 derivatives. RESULTS VNN1 overexpression on colonocytes correlates with IBD severity. VIVA mice are resistant to experimentally induced colitis. The pantetheinase activity of Vnn1 is cytoprotective in colon: it enhances CoA regeneration and metabolic adaptation of colonocytes; it favours microbiota-dependent production of short chain fatty acids and mostly butyrate, shown to regulate mucosal energetics and to be reduced in patients with IBD. This prohealing phenotype is recapitulated by treating control mice with the substrate (pantethine) or the products of pantetheinase activity prior to induction of colitis. In severe IBD, the protection conferred by the high induction of VNN1 might be compromised because its enzymatic activity may be limited by lack of available substrates. In addition, we identify the elevation of indoxyl sulfate in urine as a biomarker of Vnn1 overexpression, also detected in patients with IBD. CONCLUSION The induction of Vnn1/VNN1 during colitis in mouse and human is a compensatory mechanism to reinforce the mucosal barrier. Therefore, enhancement of vitamin B5-driven metabolism should improve mucosal healing and might increase the efficacy of anti-inflammatory therapy.
Collapse
Affiliation(s)
- Virginie Millet
- Centre d'Immunologie de Marseille Luminy, Aix Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | - Thomas Gensollen
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Maltese
- Centre d'Immunologie de Marseille Luminy, Aix Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | - Melanie Serrero
- Gastroenterology, AP-HM Hôpital Nord, Aix Marseille Université, Marseille, France
| | - Nathalie Lesavre
- Centre d'investigation Clinique (CIC), AP-HM Hôpital Nord, Aix-Marseille Université, Marseille, France
| | - Christophe Bourges
- Genetic Mechanisms of Disease Laboratory, The Francis Crick Institute, London, UK
| | - Christophe Pitaval
- Centre d'Immunologie de Marseille Luminy, Aix Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | - Sophie Cadra
- Centre d'Immunologie de Marseille Luminy, Aix Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | - Lionel Chasson
- Centre d'Immunologie de Marseille Luminy, Aix Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | - Thien Phong Vu Man
- Centre d'Immunologie de Marseille Luminy, Aix Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | - Marion Masse
- Centre d'Immunologie de Marseille Luminy, Aix Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | | | - Fabrice Tranchida
- ISM2, Aix Marseille Université, Centre National de la Recherche Scientifique, Centrale Marseille, Marseille, France
| | - Laetitia Shintu
- ISM2, Aix Marseille Université, Centre National de la Recherche Scientifique, Centrale Marseille, Marseille, France
| | - Konrad Mostert
- Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | - Erick Strauss
- Stellenbosch University, Stellenbosch, Western Cape, South Africa
| | | | | | - Achille Broggi
- Centre d'Immunologie de Marseille Luminy, Aix Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Inserm NGERE U1256, University Hospital of Nancy, University of Lorraine, Vandoeuvre-lès-Nancy, France
| | - Jean-Charles Grimaud
- Gastroenterology, AP-HM Hôpital Nord, Aix Marseille Université, Marseille, France
| | - Philippe Naquet
- Centre d'Immunologie de Marseille Luminy, Aix Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| | - Franck Galland
- Centre d'Immunologie de Marseille Luminy, Aix Marseille Université, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Marseille, France
| |
Collapse
|
21
|
Zhang X, Cong W, Lu A. Vanin-1 as a novel biomarker for chronic obstructive pulmonary disease. Heart Lung 2022; 56:91-95. [PMID: 35810677 DOI: 10.1016/j.hrtlng.2022.06.022] [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: 02/24/2022] [Revised: 06/13/2022] [Accepted: 06/23/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND The function of vascular non-inflammatory molecule 1 (Vanin-1), an enzyme essential for vitamin B5 synthesis, has not been studied yet in chronic obstructive pulmonary disease (COPD). OBJECTIVES In this study, we aimed to evaluate the expression of Vanin-1 in sera and lung tissues of COPD, and infer its possible roles in COPD. METHODS We collected blood and lung tissue specimens from 99 COPD patients and 62 non-COPD subjects. Enzyme-linked immunosorbent assay was used to determine levels of Vanin-1, pantothenic acid, interleukin-6 (IL-6), tumor necrotic factor α (TNFα), and reactive oxygen species (ROS). The receiver operating characteristics (ROC) curve was used for analysis of the diagnostic value of Vanin-1 in COPD patients. Pearson's correlation assay was used to determine the correlation between Vanin-1 and levels of inflammatory cytokines and ROS. RESULTS Vanin-1 expression was significantly higher in the sera and lung tissues of COPD patients compared to non-COPD subjects. ROC analysis showed that the area under the curve (AUC) was greater than 0.5 for both sera (AUC = 0.7342) and lung tissues (AUC = 0.9061). Pantothenic acid was also upregulated in COPD patients. IL-6, TNFα and ROS showed strong positive correlations to Vanin-1 levels in sera. CONCLUSION Vanin-1 upregulation in sera and lung tissue is a potentially valuable biomarker for COPD. Vanin-1 showed positive correlations to levels of pro-inflammatory cytokines and ROS. Together, our results support the further development of Vanin-1 as a new target for the diagnosis or treatment of COPD.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Respiratory and Intensive Medicine, Daqing Oilfield General Hospital, 9 Zhongkang Street, Daqing 163001, China
| | - Wenchao Cong
- Department of Respiratory and Intensive Medicine, Daqing Oilfield General Hospital, 9 Zhongkang Street, Daqing 163001, China
| | - Aiping Lu
- Department of Respiratory Medicine, Daqing People's Hospital, 241 Jianshe Road, Daqing 163000, China.
| |
Collapse
|
22
|
Yoneyama H, Hosohata K, Jin D, Yoshida I, Toyoda M, Kitamura I, Takai S, Usami Y. Design, synthesis, and evaluation of new vanin-1 inhibitors based on RR6. Bioorg Med Chem 2022; 65:116791. [PMID: 35537325 DOI: 10.1016/j.bmc.2022.116791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/25/2022]
Abstract
Fourteen novel vanin-1 inhibitors coded OMP-# were designed from RR6 and successfully synthesized by a nucleophilic addition-elimination reaction of the pantetheinic acid-derived Weinreb amide as a key step under Barbier conditions. The synthesized OMP compounds exhibited inhibitory activity against human serum vanin-1 in vitro. Among the synthesized compounds, OMP-7, which possesses a trifluoromethoxy group at the para-position on the phenyl ring, exhibited the most potent activity, approximately 20 times that of the mother compound RR6. OMP-7 was further subjected to an in vivo assay using a normal hamster. More potent activity was observed than that of RR6 against both serum and renal vanin-1. The activity lasted for 4 h after injection against serum vanin-1 and 1 h after injection against renal vanin-1, whereas RR6 did not show the desired activity.
Collapse
Affiliation(s)
- Hiroki Yoneyama
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1Nasahara, Takatsuki City, Osaka 569-1094, Japan
| | - Keiko Hosohata
- Education and Research Center of Clinical Pharmacy, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1Nasahara, Takatsuki City, Osaka 569-1094, Japan
| | - Denan Jin
- Department of Innovative Medicine, Graduate School of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka 569-8686, Japan
| | - Iroha Yoshida
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1Nasahara, Takatsuki City, Osaka 569-1094, Japan
| | - Miyui Toyoda
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1Nasahara, Takatsuki City, Osaka 569-1094, Japan
| | - Ikuko Kitamura
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1Nasahara, Takatsuki City, Osaka 569-1094, Japan
| | - Shinji Takai
- Department of Innovative Medicine, Graduate School of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki City, Osaka 569-8686, Japan
| | - Yoshihide Usami
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1Nasahara, Takatsuki City, Osaka 569-1094, Japan.
| |
Collapse
|
23
|
Wang G, Wang J, Du L, Li M. Visualization-Based Discovery of Vanin-1 Inhibitors for Colitis. Front Chem 2022; 9:809495. [PMID: 35155380 PMCID: PMC8831383 DOI: 10.3389/fchem.2021.809495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
The main effect of Vanin-1/VNN1 is related to its pantetheinase sulfhydrylase activity, which can hydrolyze pantetheine into pantothenic acid and cysteamine. In recent studies, the enzymatic activity of vanin-1/VNN1 has been found to be essential in the development of many diseases. The study of specific vanin-1/VNN1 inhibitors can give us a deeper understanding of its role in the disease process. In this study, different skeletal inhibitors were designed and synthesized using pyrimidine amide compounds as lead compounds. In order to screen inhibitors intuitively, a fluorescent probe PA-AFC for in vitro evaluation of inhibitors was designed and synthesized in this study, which has good sensitivity and specificity. The bioluminescent probe PA-AL was then used for cellular level and in vivo inhibitor evaluation. This screening method was convenient, economical and highly accurate. Finally, these inhibitors were applied to a mouse colitis model, confirming that vanin-1 is useful in IBD and providing a new therapeutic direction.
Collapse
|
24
|
Tian Z, Yan F, Tian X, Feng L, Cui J, Deng S, Zhang B, Xie T, Huang S, Ma X. A NIR fluorescent probe for Vanin-1 and its applications in imaging, kidney injury diagnosis, and the development of inhibitor. Acta Pharm Sin B 2022; 12:316-325. [PMID: 35127388 PMCID: PMC8799884 DOI: 10.1016/j.apsb.2021.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/22/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
Vanin-1 is an amidohydrolase that catalyses the conversion of pantetheine into the amino-thiol cysteamine and pantothenic acid (coenzyme A precursor), which plays a vital role in multiple physiological and pathological processes. In this study, an enzyme-activated near-infrared (NIR) fluorescent probe (DDAV) has been constructed for sensitively detecting Vanin-1 activity in complicated biosamples on the basis of its catalytic characteristics. DDAV exhibited a high selectivity and sensitivity toward Vanin-1 and was successfully applied to the early diagnosis of kidney injury in cisplatin-induced kidney injury model. In addition, DDAV could serve as a visual tool for in situ imaging endogenous Vanin-1 in vivo. More importantly, Enterococcus faecalis 20247 which possessed high expression of Vanin-1 was screened out from intestinal bacteria using DDAV, provided useful guidance for the rational use of NSAIDs in clinic. Finally, oleuropein as a potent natural inhibitor for Vanin-1 was discovered from herbal medicines library using a high-throughput screening method using DDAV, which held great promise for clinical therapy of inflammatory bowel disease.
Collapse
|
25
|
Casimiro-Garcia A, Allais C, Brennan A, Choi C, Dower G, Farley KA, Fleming M, Flick A, Frisbie RK, Hall J, Hepworth D, Jones H, Knafels JD, Kortum S, Lovering FE, Mathias JP, Mohan S, Morgan PM, Parng C, Parris K, Pullen N, Schlerman F, Stansfield J, Strohbach JW, Vajdos FF, Vincent F, Wang H, Wang X, Webster R, Wright SW. Discovery of a Series of Pyrimidine Carboxamides as Inhibitors of Vanin-1. J Med Chem 2021; 65:757-784. [PMID: 34967602 DOI: 10.1021/acs.jmedchem.1c01849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
A diaryl ketone series was identified as vanin-1 inhibitors from a high-throughput screening campaign. While this novel scaffold provided valuable probe 2 that was used to build target confidence, concerns over the ketone moiety led to the replacement of this group. The successful replacement of this moiety was achieved with pyrimidine carboxamides derived from cyclic secondary amines that were extensively characterized using biophysical and crystallographic methods as competitive inhibitors of vanin-1. Through optimization of potency and physicochemical and ADME properties, and guided by co-crystal structures with vanin-1, 3 was identified with a suitable profile for advancement into preclinical development.
Collapse
Affiliation(s)
- Agustin Casimiro-Garcia
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Christophe Allais
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Agnes Brennan
- Inflammation and Immunology Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Chulho Choi
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Gabriela Dower
- Inflammation and Immunology Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Kathleen A Farley
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Margaret Fleming
- Inflammation and Immunology Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Andrew Flick
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Richard K Frisbie
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Justin Hall
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - David Hepworth
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Hannah Jones
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - John D Knafels
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Steve Kortum
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Frank E Lovering
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - John P Mathias
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Sashi Mohan
- Inflammation and Immunology Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Paul M Morgan
- Inflammation and Immunology Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Chuenlei Parng
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Kevin Parris
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Nick Pullen
- Inflammation and Immunology Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Franklin Schlerman
- Inflammation and Immunology Research Unit, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - John Stansfield
- Early Clinical Development Non-Clinical Statistics, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Joseph W Strohbach
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Felix F Vajdos
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Fabien Vincent
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Hong Wang
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| | - Xiaolun Wang
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Robert Webster
- Medicine Design, Pfizer Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Stephen W Wright
- Medicine Design, Pfizer Inc., 445 Eastern Point Rd, Groton, Connecticut 06340, United States
| |
Collapse
|
26
|
Gut Microbial Metabolite-Mediated Regulation of the Intestinal Barrier in the Pathogenesis of Inflammatory Bowel Disease. Nutrients 2021; 13:nu13124259. [PMID: 34959809 PMCID: PMC8704337 DOI: 10.3390/nu13124259] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/20/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease. The disease has a multifactorial aetiology, involving genetic, microbial as well as environmental factors. The disease pathogenesis operates at the host-microbe interface in the gut. The intestinal epithelium plays a central role in IBD disease pathogenesis. Apart from being a physical barrier, the epithelium acts as a node that integrates environmental, dietary, and microbial cues to calibrate host immune response and maintain homeostasis in the gut. IBD patients display microbial dysbiosis in the gut, combined with an increased barrier permeability that contributes to disease pathogenesis. Metabolites produced by microbes in the gut are dynamic indicators of diet, host, and microbial interplay in the gut. Microbial metabolites are actively absorbed or diffused across the intestinal lining to affect the host response in the intestine as well as at systemic sites via the engagement of cognate receptors. In this review, we summarize insights from metabolomics studies, uncovering the dynamic changes in gut metabolite profiles in IBD and their importance as potential diagnostic and prognostic biomarkers of disease. We focus on gut microbial metabolites as key regulators of the intestinal barrier and their role in the pathogenesis of IBD.
Collapse
|
27
|
What role for cysteamine in the defence against infection? Emerg Top Life Sci 2021; 5:629-635. [PMID: 34027984 DOI: 10.1042/etls20200351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022]
Abstract
The aminothiol cysteamine has many potential therapeutic applications and is also an endogenous molecule, produced in the body via the activity of pantetheinase enzymes such as vanin-1. This simple small molecule is highly reactive in biological settings and much is yet unknown about its endogenous role in innate immunity to infection, including the impact of cysteamine on bacterial pathogens. We discuss the literature surrounding its biochemistry and challenges to its development as well as the multiple beneficial properties which have been uncovered that support research into its development as novel antimicrobial therapy.
Collapse
|
28
|
Xiang H, Guo F, Tao X, Zhou Q, Xia S, Deng D, Li L, Shang D. Pancreatic ductal deletion of S100A9 alleviates acute pancreatitis by targeting VNN1-mediated ROS release to inhibit NLRP3 activation. Theranostics 2021; 11:4467-4482. [PMID: 33754072 PMCID: PMC7977474 DOI: 10.7150/thno.54245] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/03/2021] [Indexed: 12/13/2022] Open
Abstract
Recent studies have proven that the overall pathophysiology of pancreatitis involves not only the pancreatic acinar cells but also duct cells, however, pancreatic duct contribution in acinar cells homeostasis is poorly known and the molecular mechanisms leading to acinar insult and acute pancreatitis (AP) are unclear. Our previous work also showed that S100A9 protein level was notably increased in AP rat pancreas through iTRAQ-based quantitative proteomic analysis. Therefore, we investigated the actions of injured duct cells on acinar cells and the S100A9-related effects and mechanisms underlying AP pathology in the present paper. Methods: In this study, we constructed S100A9 knockout (s100a9-/-) mice and an in vitro coculture system for pancreatic duct cells and acinar cells. Moreover, a variety of small molecular inhibitors of S100A9 were screened from ChemDiv through molecular docking and virtual screening methods. Results: We found that the upregulation of S100A9 induces cell injury and inflammatory response via NLRP3 activation by targeting VNN1-mediated ROS release; and loss of S100A9 decreases AP injury in vitro and in vivo. Moreover, molecular docking and mutant plasmid experiments proved that S100A9 has a direct interaction with VNN1 through the salt bridges formation of Lys57 and Glu92 residues in S100A9 protein. We further found that compounds C42H60N4O6 and C28H29F3N4O5S can significantly improve AP injury in vitro and in vivo through inhibiting S100A9-VNN1 interaction. Conclusions: Our study showed the important regulatory effect of S100A9 on pancreatic duct injury during AP and revealed that inhibition of the S100A9-VNN1 interaction may be a key therapeutic target for this disease.
Collapse
Affiliation(s)
- Hong Xiang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Fangyue Guo
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Xufeng Tao
- School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Qi Zhou
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
| | - Shilin Xia
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Dawei Deng
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Lunxu Li
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Dong Shang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116044, China
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| |
Collapse
|
29
|
Plasma Vanin-1 as a Novel Biomarker of Sepsis for Trauma Patients: A Prospective Multicenter Cohort Study. Infect Dis Ther 2021; 10:739-751. [PMID: 33624223 PMCID: PMC8116364 DOI: 10.1007/s40121-021-00414-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
Introduction Vanin-1 plays a pivotal role in oxidative stress and the inflammatory response. However, its relationship with traumatic sepsis remains unknown. The aim of our study was to evaluate whether plasma vanin-1 could be used for the early prediction of traumatic sepsis. Methods In this three-stage prospective cohort study, severe trauma patients admitted from January 2015 to October 2018 at two hospitals were enrolled. Plasma vanin-1 levels were measured by enzyme-linked immunosorbent assay (ELISA). The associations among variables and traumatic sepsis were identified by logistic regression models and the receiver operating characteristic (ROC) curve was analyzed to evaluate the diagnostic efficiency. Results A total of 426 trauma patients (22 in the discovery cohort, 283 in the internal test cohort, and 121 in the external validation cohort) and 16 healthy volunteers were recruited. The plasma vanin-1 of trauma patients was significantly higher than that of healthy volunteers (P < 0.05). Patients with sepsis had higher plasma vanin-1 than patients without sepsis in the discovery trauma cohort (P < 0.05). In the internal test cohort, plasma vanin-1 at day 1 after trauma was significantly associated with the incidence of sepsis (OR = 3.92, 95% CI 2.68–5.72, P = 1.62 × 10−12). As a predictive biomarker, vanin-1 afforded a better area under the curve (AUC) (0.82, 95% CI 0.77–0.87) than C-reaction protein (CRP) (0.62, 95% CI 0.56–0.68, P < 0.0001), procalcitonin (PCT) (0.66, 95% CI 0.60–0.71, P < 0.0001), and Acute Physiology and Chronic Health Evaluation II (APACHE II) (0.71, 95% CI 0.65–0.76, P = 6.70 × 10−3). The relevance was further validated in the external validation cohort (OR = 4.26, 95% CI 2.22–8.17, P = 1.28 × 10−5), with an AUC of 0.83 (95% CI 0.75–0.89). Vanin-1 could also improve the diagnostic efficiency of APACHE II (AUC = 0.85). Conclusions Our study demonstrated that plasma vanin-1 increased among trauma patients and was independently associated with the risk of sepsis. Vanin-1 might be a potential biomarker for the early prediction of traumatic sepsis. Trial Registration Clinicaltrials.gov Identifier, NCT01713205. Supplementary Information The online version contains supplementary material available at 10.1007/s40121-021-00414-w.
Collapse
|
30
|
Wilkerson A, Bhat N, Pham HQH, Yuksel S, Butovich I. Physiological effects of inactivation and the roles of Elovl3/ELOVL3 in maintaining ocular homeostasis. FASEB J 2021; 35:e21327. [PMID: 33455016 PMCID: PMC7891900 DOI: 10.1096/fj.202002323r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/22/2022]
Abstract
Recently, elongase of very long chain fatty acids-3 (ELOVL3) was demonstrated to play a pivotal role in physiology and biochemistry of the ocular surface by maintaining a proper balance in the lipid composition of meibum. The goal of this study was to further investigate the effects of ELOVL3 ablation in homozygous Elovl3-knockout mice (E3hom) in comparison with age and sex matched wild-type controls (E3wt). Slit lamp examination of the ocular surface of mice, and histological examination of their ocular tissues, highlighted a severe negative impact of Elovl3 inactivating mutation on the Meibomian glands (MG) and conjunctiva of mice. MG transcriptomes of the E3hom and E3wt mice were assessed and revealed a range of up- and downregulated genes related to lipid biosynthesis, inflammation, and stress response, compared with E3wt mice. Heat stage polarized light microscopy was used to assess melting characteristics of normal and abnormal meibum. The loss of Elovl3 led to a 8°C drop in the melting temperature of meibum in E3hom mice, and increased its fluidity. Also noted were the excessive accumulation of lipid material and tears around the eye and severe ocular inflammation, among other abnormalities.
Collapse
Affiliation(s)
- Amber Wilkerson
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nita Bhat
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Hoang Quoc Hai Pham
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Seher Yuksel
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Igor Butovich
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- The Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
31
|
Diel de Amorim M, Khan FA, Chenier TS, Scholtz EL, Hayes MA. Analysis of the uterine flush fluid proteome of healthy mares and mares with endometritis or fibrotic endometrial degeneration. Reprod Fertil Dev 2021; 32:572-581. [PMID: 31987068 DOI: 10.1071/rd19085] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022] Open
Abstract
The objective of this study was to evaluate the differences in the uterine flush fluid proteome between healthy mares and mares with endometritis or fibrotic endometrial degeneration (FED). Uterine flush fluid samples were collected from healthy mares (n=8; oestrus n=5 and dioestrus n=3) and mares with endometritis (n=23; oestrus n=14 and dioestrus n=9) or FED (n=7; oestrus n=6 and dioestrus n=1). Proteomic analysis was performed using label-free liquid chromatography-tandem mass spectrometry. Of 216 proteins identified during oestrus, 127 were common to all three groups, one protein was exclusively detected in healthy mares, 47 proteins were exclusively detected in mares with endometritis and four proteins were exclusively detected in mares with FED. Of 188 proteins identified during dioestrus, 113 proteins were common between healthy mares and mares with endometritis, eight proteins were exclusively detected in healthy mares and 67 proteins were exclusively detected in mares with endometritis. Quantitative analysis revealed a subset of proteins differing in abundance between the three groups during oestrus and between healthy mares and mares with endometritis during dioestrus. These results provide a springboard for evaluation of specific proteins as biomarkers of uterine health and disease and for investigation of their roles in the establishment and maintenance of pregnancy.
Collapse
Affiliation(s)
- Mariana Diel de Amorim
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada; and Present address: Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA; and Corresponding authors. Emails: ;
| | - Firdous A Khan
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada; and Department of Large Animal Medicine and Surgery, School of Veterinary Medicine, St. George's University, True Blue, St. George's, Grenada; and Present address: Department of Large Animal Medicine and Surgery, School of Veterinary Medicine, St. George's University, True Blue, St. George's, Grenada; and Corresponding authors. Emails: ;
| | - Tracey S Chenier
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada
| | - Elizabeth L Scholtz
- Department of Population Medicine, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada
| | - M Anthony Hayes
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON, N1G2W1, Canada
| |
Collapse
|
32
|
Qian J, Zhang L, Wang J, Teng Z, Cao T, Zheng L, Cao Y, Qin W, Liu Y, Guo H. Red emission ratio fluorescent probe for the activity of vanin-1 and imaging in vivo. JOURNAL OF HAZARDOUS MATERIALS 2021; 401:123863. [PMID: 33113750 DOI: 10.1016/j.jhazmat.2020.123863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/29/2020] [Accepted: 08/16/2020] [Indexed: 06/11/2023]
Abstract
Pantetheinase, also known as Vanin-1, catalyzes pantetheine to decompose into the precursor of CoA - pantothenic acid and aminothiol cysteamine. Studies have shown that Vanin-1 plays an important role in many important physiological pathologies. In this paper, a new red emission ratio fluorescent probe DCM-PA (I640 nm/I564 nm) has been implemented to detect the activity of Vanin-1 in cells and vivo. DCM-PA has short response time (30 min), high selectivity and low sensitivity (DL =0.69 ng/mL). Also, we have applied DCM-PA for imaging in cells and mice, and the results have indicated that the probe has a non-negligible potential for monitoring the activity of Vanin-1 in situ, benefiting further to study the role of Vanin-1 in physiology and pathology. In addition, the up-regulation of this enzyme by starvation confirmed the inevitable connection between diabetes and abnormal expression of Vanin-1.
Collapse
Affiliation(s)
- Jing Qian
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and Key Laboratory of Special Function Materials and Structure Design (MOE), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Liang Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, PR China
| | - Jiemin Wang
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and Key Laboratory of Special Function Materials and Structure Design (MOE), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Zhidong Teng
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu Province 730046, PR China
| | - Ting Cao
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and Key Laboratory of Special Function Materials and Structure Design (MOE), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Lei Zheng
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and Key Laboratory of Special Function Materials and Structure Design (MOE), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Yuping Cao
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and Key Laboratory of Special Function Materials and Structure Design (MOE), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Wenwu Qin
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and Key Laboratory of Special Function Materials and Structure Design (MOE), College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China.
| | - Yun Liu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, PR China.
| | - Huichen Guo
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu Province 730046, PR China.
| |
Collapse
|
33
|
Unterschemmann K, Ehrmann A, Herzig I, Andreevski AL, Lustig K, Schmeck C, Eitner F, Grundmann M. Pharmacological inhibition of Vanin-1 is not protective in models of acute and chronic kidney disease. Am J Physiol Renal Physiol 2021; 320:F61-F73. [PMID: 33196323 DOI: 10.1152/ajprenal.00373.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/08/2020] [Indexed: 12/31/2022] Open
Abstract
Oxidative stress is a key concept in basic, translational, and clinical research to understand the pathophysiology of various disorders, including cardiovascular and renal diseases. Although attempts to directly reduce oxidative stress with redox-active substances have until now largely failed to prove clinical benefit, indirect approaches to combat oxidative stress enzymatically have gained further attention as potential therapeutic strategies. The pantetheinase Vanin-1 is expressed on kidney proximal tubular cells, and its reaction product cysteamine is described to negatively affect redox homeostasis by inhibiting the replenishment of cellular antioxidative glutathione stores. Vanin-1-deficient mice were shown to be protected against oxidative stress damage. The aim of this study was to elucidate whether pharmacological inhibition of Vanin-1 protects mice from oxidative stress-related acute or chronic kidney injury as well. By studying renal ischemia-reperfusion injury in Col4α3-/- (Alport syndrome) mice and in vitro hypoxia-reoxygenation in human proximal tubular cells we found that treatment with a selective and potent Vanin-1 inhibitor resulted in ample inhibition of enzymatic activity in vitro and in vivo. However, surrogate parameters of metabolic and redox homeostasis were only partially and insufficiently affected. Consequently, apoptosis and reactive oxygen species level in tubular cells as well as overall kidney function and fibrotic processes were not improved by Vanin-1 inhibition. We thus conclude that Vanin-1 functionality in the context of cardiovascular diseases needs further investigation and the biological relevance of pharmacological Vanin-1 inhibition for the treatment of kidney diseases remains to be proven.
Collapse
MESH Headings
- Acute Kidney Injury/enzymology
- Acute Kidney Injury/genetics
- Acute Kidney Injury/pathology
- Acute Kidney Injury/prevention & control
- Amidohydrolases/antagonists & inhibitors
- Amidohydrolases/genetics
- Amidohydrolases/metabolism
- Animals
- Apoptosis/drug effects
- Autoantigens/genetics
- Autoantigens/metabolism
- Cell Line
- Collagen Type IV/genetics
- Collagen Type IV/metabolism
- Disease Models, Animal
- Enzyme Inhibitors/pharmacokinetics
- Enzyme Inhibitors/pharmacology
- Fibrosis
- GPI-Linked Proteins/antagonists & inhibitors
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Humans
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/enzymology
- Kidney Tubules, Proximal/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Nephritis, Hereditary/enzymology
- Nephritis, Hereditary/genetics
- Nephritis, Hereditary/pathology
- Nephritis, Hereditary/prevention & control
- Oxidative Stress/drug effects
- Renal Insufficiency, Chronic/enzymology
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/prevention & control
- Reperfusion Injury/enzymology
- Reperfusion Injury/genetics
- Reperfusion Injury/pathology
- Reperfusion Injury/prevention & control
- Mice
Collapse
Affiliation(s)
| | | | - Ina Herzig
- Drug Discovery Sciences, Bayer Pharmaceuticals, Wuppertal, Germany
| | | | - Klemens Lustig
- Research and Early Development, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Carsten Schmeck
- Drug Discovery Sciences, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Frank Eitner
- Research and Early Development, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Manuel Grundmann
- Research and Early Development, Bayer Pharmaceuticals, Wuppertal, Germany
| |
Collapse
|
34
|
Gurung AB, Bhutia JT, Bhattacharjee A. High-throughput virtual screening of novel potent inhibitor(s) for Human Vanin-1 enzyme. J Biomol Struct Dyn 2020; 40:4208-4223. [PMID: 33289461 DOI: 10.1080/07391102.2020.1854857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Vanin-1 (VNN1) is a glycosylphosphatidylinositol (GPI)-anchored ectoenzyme which hydrolyzes pantetheine to pantothenic acid and cysteamine. It has emerged as a promising drug target for many human diseases associated with oxidative stress and inflammatory pathways. In the present study we used structure-based virtual screening approach for the identification of small molecule inhibitors of vanin-1. A chemical library consisting of natural compounds, synthetic compounds and RRV analogs were screened for drug-like molecules. The filtered molecules were subjected to molecular docking studies. Three potential hits-ZINC04073864 (Natural compound), CID227017 (synthetic compound) and CID129558381 (RRV analog)-were identified for the target enzyme. The molecules form good number of hydrogen bonds with the catalytic residues such as Glu79, Lys178 and Cys211. The apo-VNN1 and VNN1-ligand complexes were subjected to molecular dynamics (MD) simulation for 30 ns. The geometric properties such as root mean square deviation, radius of gyration, solvent accessible surface area, number of hydrogen bonds and the distance between the catalytic triad residues-Glu79, Lys178 and Cys211 were altered upon binding of the compounds. Essential dynamics and entropic studies further confirmed that the fluctuations in VNN1 decrease upon binding of the compounds. The lead molecules were stable throughout the simulation time period. Molecular Mechanics Poisson-Boltzmann Surface Area (MM/PBSA) studies showed that Van der Waals interaction energy contributes significantly to the total binding free energy. Thus, our study reveals three lead molecules-ZINC04073864, CID227017 and CID129558381 as potential inhibitors of Vanin-1 which can be validated through further studies. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Arun Bahadur Gurung
- Computational Biology Laboratory, Department of Biotechnology and Bioinformatics, North Eastern Hill University, Shillong, Meghalaya, India.,Department of Basic Sciences and Social Sciences, North-Eastern Hill University, Shillong, Meghalaya, India
| | - Jigmi Tshering Bhutia
- Computational Biology Laboratory, Department of Biotechnology and Bioinformatics, North Eastern Hill University, Shillong, Meghalaya, India
| | - Atanu Bhattacharjee
- Computational Biology Laboratory, Department of Biotechnology and Bioinformatics, North Eastern Hill University, Shillong, Meghalaya, India
| |
Collapse
|
35
|
Czumaj A, Szrok-Jurga S, Hebanowska A, Turyn J, Swierczynski J, Sledzinski T, Stelmanska E. The Pathophysiological Role of CoA. Int J Mol Sci 2020; 21:ijms21239057. [PMID: 33260564 PMCID: PMC7731229 DOI: 10.3390/ijms21239057] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
The importance of coenzyme A (CoA) as a carrier of acyl residues in cell metabolism is well understood. Coenzyme A participates in more than 100 different catabolic and anabolic reactions, including those involved in the metabolism of lipids, carbohydrates, proteins, ethanol, bile acids, and xenobiotics. However, much less is known about the importance of the concentration of this cofactor in various cell compartments and the role of altered CoA concentration in various pathologies. Despite continuous research on these issues, the molecular mechanisms in the regulation of the intracellular level of CoA under pathological conditions are still not well understood. This review summarizes the current knowledge of (a) CoA subcellular concentrations; (b) the roles of CoA synthesis and degradation processes; and (c) protein modification by reversible CoA binding to proteins (CoAlation). Particular attention is paid to (a) the roles of changes in the level of CoA under pathological conditions, such as in neurodegenerative diseases, cancer, myopathies, and infectious diseases; and (b) the beneficial effect of CoA and pantethine (which like CoA is finally converted to Pan and cysteamine), used at pharmacological doses for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Aleksandra Czumaj
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdańsk, Poland;
| | - Sylwia Szrok-Jurga
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (A.H.); (J.T.)
| | - Areta Hebanowska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (A.H.); (J.T.)
| | - Jacek Turyn
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (A.H.); (J.T.)
| | - Julian Swierczynski
- State School of Higher Vocational Education in Koszalin, 75-582 Koszalin, Poland;
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-211 Gdańsk, Poland;
- Correspondence: (T.S.); (E.S.); Tel.: +48-(0)-583-491-479 (T.S.)
| | - Ewa Stelmanska
- Department of Biochemistry, Faculty of Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (S.S.-J.); (A.H.); (J.T.)
- Correspondence: (T.S.); (E.S.); Tel.: +48-(0)-583-491-479 (T.S.)
| |
Collapse
|
36
|
B Vitamins and Their Role in Immune Regulation and Cancer. Nutrients 2020; 12:nu12113380. [PMID: 33158037 PMCID: PMC7693142 DOI: 10.3390/nu12113380] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
B group vitamins represent essential micronutrients for myriad metabolic and regulatory processes required for human health, serving as cofactors used by hundreds of enzymes that carry out essential functions such as energy metabolism, DNA and protein synthesis and other critical functions. B vitamins and their corresponding vitamers are universally essential for all cellular life forms, from bacteria to humans. Humans are unable to synthesize most B vitamins and are therefore dependent on their diet for these essential micronutrients. More recently, another source of B vitamins has been identified which is derived from portions of the 1013 bacterial cells inhabiting the gastrointestinal tract. Here we review the expanding literature examining the relationship between B vitamins and the immune system and diverse cancers. Evidence of B vitamin’s role in immune cell regulation has accumulated in recent years and may help to clarify the disparate findings of numerous studies attempting to link B vitamins to cancer development. Much work remains to be carried out to fully clarify these relationships as the complexity of B vitamins’ essential functions complicates an unequivocal assessment of their beneficial or detrimental effects in inflammation and cancers.
Collapse
|
37
|
Qian J, Teng Z, Wang J, Zhang L, Cao T, Zheng L, Cao Y, Qin W, Liu Y, Guo H. Visible to Near-Infrared Emission Ratiometric Fluorescent Probe for the Detection of Vanin-1 In Vivo. ACS Sens 2020; 5:2806-2813. [PMID: 32786380 DOI: 10.1021/acssensors.0c00880] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pantetheinase (Vanin-1) is an ectoenzyme, which involves the metabolic pathway of coenzyme A (CoA), and can decompose pantetheine into pantothenic acid (CoA precursor) and aminothiol cysteamine. Previous studies have revealed that Vanin-1 with essential biological functions is closely related to many diseases. However, the lack of simple and effective detection methods has severely hindered the further study of Vanin-1's physiological functions. In this work, we have developed a near-infrared (NIR) emission ratio fluorescent probe TMN-PA (I645 nm/I568 nm) that enables us to detect Vanin-1 rapidly (in 15 min) with a minimum detection limit of 0.37 ng/mL. What is more, this probe shows excellent potential in in situ real-time monitoring of the endogenous Vanin-1, contributing to further research on Vanin-1 and understanding its mechanisms in physiological pathology. To our knowledge, this probe is the first NIR emission ratio (I645 nm/I568 nm) fluorescent probe ever reported to monitor the activity of Vanin-1 in vivo.
Collapse
Affiliation(s)
- Jing Qian
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Special Function Materials and Structure Design (MOF), and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Zhidong Teng
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu Province 730046, P. R. China
| | - Jiemin Wang
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Special Function Materials and Structure Design (MOF), and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Liang Zhang
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, P. R. China
| | - Ting Cao
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Special Function Materials and Structure Design (MOF), and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Lei Zheng
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Special Function Materials and Structure Design (MOF), and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Yuping Cao
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Special Function Materials and Structure Design (MOF), and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Wenwu Qin
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Special Function Materials and Structure Design (MOF), and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, P. R. China
| | - Yun Liu
- Department of Radiology, Xinqiao Hospital, Army Medical University, Chongqing 400037, P. R. China
| | - Huichen Guo
- State Key Laboratory of Veterinary Etiological Biology and Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Lanzhou, Gansu Province 730046, P. R. China
| |
Collapse
|
38
|
Chen S, Zhang W, Sun C, Song M, Liu S, Xu M, Zhang X, Liu L, Liu C. Systemic Nanoparticle-Mediated Delivery of Pantetheinase Vanin-1 Regulates Lipolysis and Adiposity in Abdominal White Adipose Tissue. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000542. [PMID: 32714762 PMCID: PMC7375228 DOI: 10.1002/advs.202000542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/12/2020] [Indexed: 05/04/2023]
Abstract
Lipolysis in white adipose tissue (WAT) occurs in response to nutritional signals and helps to regulate lipid turnover/adiposity in animals. However, the causal relationships and the mechanisms controlling WAT morphology are not clear. In this report, Vanin-1, a pantetheinase, is shown to be a novel determinant for lipolysis and adiposity. The expression of Vanin-1 in the abdominal WAT is positively correlated with lipolysis both in mice and in humans. Mice with global Vanin-1 deficiency exhibit adipocyte hypertrophy and impaired lipolysis. Use of a nanosystem comprising P3-peptide, chitosan oligosaccharide lactate, and polyethylene glycol that controls Vanin-1 expression in the abdominal WAT shows that WAT-specific Vanin-1 knockdown blocks fasting-induced lipolysis and prevents WAT loss. However, WAT-specific Vanin-1 mRNA restoration rescues impaired lipolysis and improves glucose/insulin intolerance in diabetic db/db mice. Mechanistically, Vanin-1 induces PPARγ activity and subsequently facilitates its activation on the proximal promoters of lipolytic genes. Thus, an essential role of Vanin-1 in the regulation of lipolysis and adiposity is revealed, and a functional RNA delivering strategy for specific intervention of Vanin-1 expression in WAT is shown. These findings provide a promising approach to treat metabolic diseases caused by dysregulation of Vanin-1 and lipolysis.
Collapse
Affiliation(s)
- Siyu Chen
- State Key Laboratory of Natural Medicines, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjing211198China
| | - Wenxiang Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjing211198China
| | - Chen Sun
- State Key Laboratory of Natural Medicines, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjing211198China
| | - Mingming Song
- State Key Laboratory of Natural Medicines, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjing211198China
| | - Shuang Liu
- State Key Laboratory of Natural Medicines, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjing211198China
| | - Mengyi Xu
- State Key Laboratory of Natural Medicines, School of Life Science and TechnologyChina Pharmaceutical UniversityNanjing211198China
| | - Xiaojin Zhang
- Department of GeriatricsFirst Affiliated Hospital with Nanjing Medical UniversityNanjing210029China
| | - Li Liu
- Department of GeriatricsFirst Affiliated Hospital with Nanjing Medical UniversityNanjing210029China
| | - Chang Liu
- State key Laboratory of Pharmaceutical BiotechnologyNanjing UniversityNanjing210046China
| |
Collapse
|
39
|
Wang Z, Yu J, Hua N, Li J, Xu L, Yao W, Gu Z. Regulation of chicken vanin1 gene expression by peroxisome proliferators activated receptor α and miRNA-181a-5p. Anim Biosci 2020; 34:172-184. [PMID: 32299163 PMCID: PMC7876720 DOI: 10.5713/ajas.19.1000] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/30/2020] [Indexed: 11/27/2022] Open
Abstract
Objective Vanin1 (VNN1) is a pantetheinase that can catalyze the hydrolysis of pantetheine to produce pantothenic acid and cysteamine. Our previous studies showed that VNN1 is specifically expressed in chicken liver. In this study, we aimed to investigate the roles of peroxisome proliferators activated receptor α (PPARα) and miRNA-181a-5p in regulating VNN1 gene expression in chicken liver. Methods 5′-RACE was performed to identify the transcription start site of chicken VNN1. JASPAR and TFSEARCH were used to analyze the potential transcription factor binding sites in the promoter region of chicken VNN1 and miRanda was used to search miRNA binding sites in 3′ untranslated region (3′UTR) of chicken VNN1. We used a knock-down strategy to manipulate PPARα (or miRNA-181a-5p) expression levels in vitro to further investigate its effect on VNN1 gene transcription. Luciferase reporter assays were used to explore the specific regions of VNN1 targeted by PPARα and miRNA-181a-5p. Results Sequence analysis of the VNN1 promoter region revealed several transcription factor-binding sites, including hepatocyte nuclear factor 1α (HNF1α), PPARα, and CCAAT/enhancer binding protein α. GW7647 (a specific agonist of PPARα) increased the expression level of VNN1 mRNA in chicken primary hepatocytes, whereas knockdown of PPARα with siRNA increased VNN1 mRNA expression. Moreover, the predicted PPARα-binding site was confirmed to be necessary for PPARα regulation of VNN1 gene expression. In addition, the VNN1 3′UTR contains a sequence that is completely complementary to nucleotides 1 to 7 of miRNA-181a-5p. Overexpression of miR-181a-5p significantly decreased the expression level of VNN1 mRNA. Conclusion This study demonstrates that PPARα is an important transcriptional activator of VNN1 gene expression and that miRNA-181a-5p acts as a negative regulator of VNN1 expression in chicken hepatocytes.
Collapse
Affiliation(s)
- Zhongliang Wang
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu, 215500, Jiangsu, China.,College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Jianfeng Yu
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu, 215500, Jiangsu, China
| | - Nan Hua
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu, 215500, Jiangsu, China
| | - Jie Li
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu, 215500, Jiangsu, China.,College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Lu Xu
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu, 215500, Jiangsu, China
| | - Wen Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Zhiliang Gu
- School of Biology and Food Engineering, Changshu Institute of Technology, Changshu, 215500, Jiangsu, China
| |
Collapse
|
40
|
Naquet P, Kerr EW, Vickers SD, Leonardi R. Regulation of coenzyme A levels by degradation: the 'Ins and Outs'. Prog Lipid Res 2020; 78:101028. [PMID: 32234503 DOI: 10.1016/j.plipres.2020.101028] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/09/2020] [Accepted: 02/22/2020] [Indexed: 02/06/2023]
Abstract
Coenzyme A (CoA) is the predominant acyl carrier in mammalian cells and a cofactor that plays a key role in energy and lipid metabolism. CoA and its thioesters (acyl-CoAs) regulate a multitude of metabolic processes at different levels: as substrates, allosteric modulators, and via post-translational modification of histones and other non-histone proteins. Evidence is emerging that synthesis and degradation of CoA are regulated in a manner that enables metabolic flexibility in different subcellular compartments. Degradation of CoA occurs through distinct intra- and extracellular pathways that rely on the activity of specific hydrolases. The pantetheinase enzymes specifically hydrolyze pantetheine to cysteamine and pantothenate, the last step in the extracellular degradation pathway for CoA. This reaction releases pantothenate in the bloodstream, making this CoA precursor available for cellular uptake and de novo CoA synthesis. Intracellular degradation of CoA depends on specific mitochondrial and peroxisomal Nudix hydrolases. These enzymes are also active against a subset of acyl-CoAs and play a key role in the regulation of subcellular (acyl-)CoA pools and CoA-dependent metabolic reactions. The evidence currently available indicates that the extracellular and intracellular (acyl-)CoA degradation pathways are regulated in a coordinated and opposite manner by the nutritional state and maximize the changes in the total intracellular CoA levels that support the metabolic switch between fed and fasted states in organs like the liver. The objective of this review is to update the contribution of these pathways to the regulation of metabolism, physiology and pathology and to highlight the many questions that remain open.
Collapse
Affiliation(s)
- Philippe Naquet
- Aix Marseille Univ, INSERM, CNRS, Centre d'Immunologie de Marseille-Luminy, Marseille, France.
| | - Evan W Kerr
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States of America
| | - Schuyler D Vickers
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States of America
| | - Roberta Leonardi
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States of America.
| |
Collapse
|
41
|
Gheita AA, Gheita TA, Kenawy SA. The potential role of B5: A stitch in time and switch in cytokine. Phytother Res 2019; 34:306-314. [DOI: 10.1002/ptr.6537] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/23/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Alaa A. Gheita
- Plastic Surgery Department, Faculty of MedicineCairo University Cairo Egypt
- Egyptian Society of Plastic and Reconstructive Surgeons Egypt
| | - Tamer A. Gheita
- Rheumatology and Clinical Immunology Department, Faculty of MedicineCairo University Cairo Egypt
- Graduate Studies and Research AffairCairo University Cairo Egypt
| | - Sanaa A. Kenawy
- Pharmacology Department, Faculty of PharmacyCairo University Cairo Egypt
- Royal College of SurgeonsLondon University London UK
| |
Collapse
|
42
|
Bartucci R, Salvati A, Olinga P, Boersma YL. Vanin 1: Its Physiological Function and Role in Diseases. Int J Mol Sci 2019; 20:E3891. [PMID: 31404995 PMCID: PMC6719204 DOI: 10.3390/ijms20163891] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/06/2019] [Accepted: 08/08/2019] [Indexed: 12/11/2022] Open
Abstract
The enzyme vascular non-inflammatory molecule-1 (vanin 1) is highly expressed at gene and protein level in many organs, such as the liver, intestine, and kidney. Its major function is related to its pantetheinase activity; vanin 1 breaks down pantetheine in cysteamine and pantothenic acid, a precursor of coenzyme A. Indeed, its physiological role seems strictly related to coenzyme A metabolism, lipid metabolism, and energy production. In recent years, many studies have elucidated the role of vanin 1 under physiological conditions in relation to oxidative stress and inflammation. Vanin's enzymatic activity was found to be of key importance in certain diseases, either for its protective effect or as a sensitizer, depending on the diseased organ. In this review, we discuss the role of vanin 1 in the liver, kidney, intestine, and lung under physiological as well as pathophysiological conditions. Thus, we provide a more complete understanding and overview of its complex function and contribution to some specific pathologies.
Collapse
Affiliation(s)
- Roberta Bartucci
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Division of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Division of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Anna Salvati
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Peter Olinga
- Division of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Ykelien L Boersma
- Division of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
43
|
Braitsch CM, Azizoglu DB, Htike Y, Barlow HR, Schnell U, Chaney CP, Carroll TJ, Stanger BZ, Cleaver O. LATS1/2 suppress NFκB and aberrant EMT initiation to permit pancreatic progenitor differentiation. PLoS Biol 2019; 17:e3000382. [PMID: 31323030 PMCID: PMC6668837 DOI: 10.1371/journal.pbio.3000382] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 07/31/2019] [Accepted: 07/02/2019] [Indexed: 12/25/2022] Open
Abstract
The Hippo pathway directs cell differentiation during organogenesis, in part by restricting proliferation. How Hippo signaling maintains a proliferation-differentiation balance in developing tissues via distinct molecular targets is only beginning to be understood. Our study makes the unexpected finding that Hippo suppresses nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) signaling in pancreatic progenitors to permit cell differentiation and epithelial morphogenesis. We find that pancreas-specific deletion of the large tumor suppressor kinases 1 and 2 (Lats1/2PanKO) from mouse progenitor epithelia results in failure to differentiate key pancreatic lineages: acinar, ductal, and endocrine. We carried out an unbiased transcriptome analysis to query differentiation defects in Lats1/2PanKO. This analysis revealed increased expression of NFκB activators, including the pantetheinase vanin1 (Vnn1). Using in vivo and ex vivo studies, we show that VNN1 activates a detrimental cascade of processes in Lats1/2PanKO epithelium, including (1) NFκB activation and (2) aberrant initiation of epithelial-mesenchymal transition (EMT), which together disrupt normal differentiation. We show that exogenous stimulation of VNN1 or NFκB can trigger this cascade in wild-type (WT) pancreatic progenitors. These findings reveal an unexpected requirement for active suppression of NFκB by LATS1/2 during pancreas development, which restrains a cell-autonomous deleterious transcriptional program and thereby allows epithelial differentiation.
Collapse
Affiliation(s)
- Caitlin M. Braitsch
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - D. Berfin Azizoglu
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Yadanar Htike
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Haley R. Barlow
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ulrike Schnell
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Christopher P. Chaney
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Thomas J. Carroll
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ben Z. Stanger
- Department of Medicine and Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ondine Cleaver
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
44
|
Yoshii K, Hosomi K, Sawane K, Kunisawa J. Metabolism of Dietary and Microbial Vitamin B Family in the Regulation of Host Immunity. Front Nutr 2019; 6:48. [PMID: 31058161 PMCID: PMC6478888 DOI: 10.3389/fnut.2019.00048] [Citation(s) in RCA: 335] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/01/2019] [Indexed: 12/28/2022] Open
Abstract
Vitamins are micronutrients that have physiological effects on various biological responses, including host immunity. Therefore, vitamin deficiency leads to increased risk of developing infectious, allergic, and inflammatory diseases. Since B vitamins are synthesized by plants, yeasts, and bacteria, but not by mammals, mammals must acquire B vitamins from dietary or microbial sources, such as the intestinal microbiota. Similarly, some intestinal bacteria are unable to synthesize B vitamins and must acquire them from the host diet or from other intestinal bacteria for their growth and survival. This suggests that the composition and function of the intestinal microbiota may affect host B vitamin usage and, by extension, host immunity. Here, we review the immunological functions of B vitamins and their metabolism by intestinal bacteria with respect to the control of host immunity.
Collapse
Affiliation(s)
- Ken Yoshii
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Kento Sawane
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Innovation Center, Nippon Flour Mills Co., Ltd., Atsugi, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Graduate School of Dentistry, Osaka University, Osaka, Japan
- Department of Microbiology and Immunology, Graduate School of Medicine, Kobe University, Hyogo, Japan
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
45
|
Ha M, Jeong H, Roh JS, Lee B, Lee D, Han ME, Oh SO, Sohn DH, Kim YH. VNN3 is a potential novel biomarker for predicting prognosis in clear cell renal cell carcinoma. Anim Cells Syst (Seoul) 2019; 23:112-117. [PMID: 30949398 PMCID: PMC6440501 DOI: 10.1080/19768354.2019.1583126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/13/2019] [Accepted: 02/10/2019] [Indexed: 02/07/2023] Open
Abstract
Although pathological observations provide approximate prognoses, it is difficult to achieve prognosis in patients with existing prognostic factors. Therefore, it is very important to find appropriate biomarkers to achieve accurate cancer prognosis. Renal cell carcinoma (RCC) has several subtypes, the discrimination of which is crucial for proper treatment. Here, we present a novel biomarker, VNN3, which is used to prognose clear cell renal cell carcinoma (ccRCC), the most common and aggressive subtype of kidney cancer. Patient information analyzed in our study was extracted from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) cohorts. VNN3 expression was considerably higher in stages III and IV than in stages I and II. Moreover, Kaplan–Meier curves associated high VNN3 expression with poor prognoses (TCGA, p < .0001; ICGC, p = .00076), confirming that ccRCC prognosis can be predicted via VNN3 expression patterns. Consistent with all patient results, the prognosis of patients with higher VNN3 expression was worse in both low stage (I and II) and high stage (III and IV) (TCGA, p < 0.0001 in stage I and II; ICGC, p = 0.028 in stage I and II; TCGA, p = 0.005 in stage III and IV). Area under the curve and receiver operating characteristic curves supported our results that highlighted VNN3 expression as a suitable ccRCC biomarker. Multivariate analysis also verified the prognostic performance of VNN3 expression (TCGA, p < .001; ICGC, p = .017). Altogether, we suggest that VNN3 is applicable as a new biomarker to establish prognosis in patients with ccRCC.
Collapse
Affiliation(s)
- Mihyang Ha
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Hoim Jeong
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jong Seong Roh
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Beomgu Lee
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Myoung-Eun Han
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Department of Biomedical Informatics, Pusan National University School of Medicine, Yangsan, Republic of Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| |
Collapse
|
46
|
Protein expression in the liver and blood serum in chickens in response to Salmonella Enteritidis infection. Vet Immunol Immunopathol 2018; 205:10-16. [PMID: 30458997 DOI: 10.1016/j.vetimm.2018.10.006] [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: 07/26/2018] [Revised: 10/03/2018] [Accepted: 10/17/2018] [Indexed: 01/20/2023]
Abstract
Events occurring in the chicken caecum following Salmonella Enteritidis infection are relatively well-described. However, mechanisms of the immune response and defence beyond the intestinal tract are less well-described. In this study, we therefore determined changes in protein abundance in the liver and blood serum in response to S. Enteritidis infection using the unbiased approach of shotgun proteomics. Complement and coagulation cascades, TNF signalling, antigen processing and presentation was activated in the liver following infection with S. Enteritidis. Chicken proteins that decreased in the liver were involved in glycolysis, the citrate cycle, oxidative phosphorylation and fatty acid metabolism. No functional category was significantly activated or suppressed in the serum. Concerning individual proteins, VNN1, SAA, AVD, SERPINA3, SERPINB10, AGT, MRP126 or CP increased in abundance both in the liver and serum. MT4, MT3, PTGDS, GLRX and TGM4, though highly inducible in the liver, did not increase in the serum. PIGR, SERPINF2 and IGJ increased in the serum but not in the liver. SERPINA4, apoAIV, CLEC3B, SERPINF1, HRG, AHSG and ALB decreased both in the liver and serum. Avidin-like LOC431660, THRSP, GATM, GGACT, ACOX1, ALDOB or FABP7 decreased in the liver but not in the serum. Finally, CKM, CKB, PLTP, COMP, IGFALS, AMY1A or SERPIND1 decreased in the serum after S. Enteritidis infection but not in the liver. Differently abundant proteins characterise the chicken's response to infection and can be also used as markers of chicken health status.
Collapse
|
47
|
Giessner C, Millet V, Mostert KJ, Gensollen T, Vu Manh TP, Garibal M, Dieme B, Attaf-Bouabdallah N, Chasson L, Brouilly N, Laprie C, Lesluyes T, Blay JY, Shintu L, Martin JC, Strauss E, Galland F, Naquet P. Vnn1 pantetheinase limits the Warburg effect and sarcoma growth by rescuing mitochondrial activity. Life Sci Alliance 2018; 1:e201800073. [PMID: 30456364 PMCID: PMC6238586 DOI: 10.26508/lsa.201800073] [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: 04/23/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022] Open
Abstract
Expression of the Vnn1 pantetheinase by sarcomas is tumor suppressive by limiting the use of aerobic glycolysis for growth and rescuing mitochondrial activity through CoA regeneration. Like other tumors, aggressive soft tissue sarcomas (STS) use glycolysis rather than mitochondrial oxidative phosphorylation (OXPHOS) for growth. Given the importance of the cofactor coenzyme A (CoA) in energy metabolism, we investigated the impact of Vnn1 pantetheinase—an enzyme that degrades pantetheine into pantothenate (vitamin B5, the CoA biosynthetic precursor) and cysyteamine—on tumor growth. Using two models, we show that Vnn1+ STS remain differentiated and grow slowly, and that in patients a detectable level of VNN1 expression in STS is associated with an improved prognosis. Increasing pantetheinase activity in aggressive tumors limits their growth. Using combined approaches, we demonstrate that Vnn1 permits restoration of CoA pools, thereby maintaining OXPHOS. The simultaneous production of cysteamine limits glycolysis and release of lactate, resulting in a partial inhibition of STS growth in vitro and in vivo. We propose that the Warburg effect observed in aggressive STS is reversed by induction of Vnn1 pantetheinase and the rewiring of cellular energy metabolism by its products.
Collapse
Affiliation(s)
- Caroline Giessner
- Aix Marseille Univ, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Marseille, France
| | - Virginie Millet
- Aix Marseille Univ, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Marseille, France
| | - Konrad J Mostert
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Thomas Gensollen
- Aix Marseille Univ, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Marseille, France
| | - Thien-Phong Vu Manh
- Aix Marseille Univ, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Marseille, France
| | - Marc Garibal
- Aix Marseille Univ, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, C2VN, Marseille, France
| | - Binta Dieme
- Aix Marseille Univ, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, C2VN, Marseille, France
| | - Noudjoud Attaf-Bouabdallah
- Aix Marseille Univ, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Marseille, France
| | - Lionel Chasson
- Aix Marseille Univ, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Marseille, France
| | - Nicolas Brouilly
- Aix Marseille Univ, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut de Biologie de Développement de Marseille, Marseille, France
| | | | - Tom Lesluyes
- Centre Lyon Bérard, Université Claude Bernard, Lyon 1, Lyon Recherche Innovation contre le Cancer, Lyon, France
| | - Jean Yves Blay
- Centre Lyon Bérard, Université Claude Bernard, Lyon 1, Lyon Recherche Innovation contre le Cancer, Lyon, France
| | - Laetitia Shintu
- Aix Marseille Université, Centre National de la Recherche Scientifique, Centrale Marseille, ISM2, Marseille, France
| | - Jean Charles Martin
- Aix Marseille Univ, Institut National de la Santé et de la Recherche Médicale, Institut National de la Recherche Agronomique, C2VN, Marseille, France
| | - Erick Strauss
- Department of Biochemistry, Stellenbosch University, Stellenbosch, South Africa
| | - Franck Galland
- Aix Marseille Univ, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Marseille, France
| | - Philippe Naquet
- Aix Marseille Univ, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Centre d'Immunologie de Marseille Luminy, Marseille, France
| |
Collapse
|
48
|
Lin Y, Gao Y, Ma Z, Li Z, Tang C, Qin X, Zhang Z, Wang G, Du L, Li M. Bioluminescent Probe for Detection of Starvation-Induced Pantetheinase Upregulation. Anal Chem 2018; 90:9545-9550. [PMID: 29976064 DOI: 10.1021/acs.analchem.8b02266] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Pantetheinase, a glycosylphosphatidylinositol (GPI) anchored enzyme, overexpresses in intestine, liver, and kidney with various biological functions such as its linkage to the inflammation and some metabolic diseases. It can hydrolyze pantetheine to cysteamine, an antioxidant, and pantothenic acid (Vitamin B5) that is an essential component of coenzyme A (CoA). Until now, very few analytic methods were developed for this enzyme, hampering the further investigation of its biological functions. In this work, we report the design, synthesis, and biological examination of a highly sensitive bioluminogenic probe for pantetheinase with a limit of detection of 1.14 ng/mL. Furthermore, animal experiments validated that our probe can be applied to detect the endogenous pantetheinase activity. To the best of our knowledge, this is the first bioluminogenic probe achieving the detection of pantetheinase level in vivo.
Collapse
Affiliation(s)
- Yuxing Lin
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy , Shandong University , Jinan , Shandong 250012 , China
| | - Yuqi Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy , Shandong University , Jinan , Shandong 250012 , China
| | - Zhao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy , Shandong University , Jinan , Shandong 250012 , China
| | - Zhenzhen Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy , Shandong University , Jinan , Shandong 250012 , China
| | - Chunchao Tang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy , Shandong University , Jinan , Shandong 250012 , China
| | - Xiaojun Qin
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy , Shandong University , Jinan , Shandong 250012 , China
| | - Zheng Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy , Shandong University , Jinan , Shandong 250012 , China
| | - Guankai Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy , Shandong University , Jinan , Shandong 250012 , China
| | - Lupei Du
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy , Shandong University , Jinan , Shandong 250012 , China
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy , Shandong University , Jinan , Shandong 250012 , China.,State Key Laboratory of Microbial Technology , Shandong University , Jinan , Shandong 250100 , China
| |
Collapse
|
49
|
Trivedi PJ, Tickle J, Vesterhus MN, Eddowes PJ, Bruns T, Vainio J, Parker R, Smith D, Liaskou E, Thorbjørnsen LW, Hirschfield GM, Auvinen K, Hubscher SG, Salmi M, Adams DH, Weston CJ. Vascular adhesion protein-1 is elevated in primary sclerosing cholangitis, is predictive of clinical outcome and facilitates recruitment of gut-tropic lymphocytes to liver in a substrate-dependent manner. Gut 2018; 67:1135-1145. [PMID: 28428344 PMCID: PMC5969351 DOI: 10.1136/gutjnl-2016-312354] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 03/23/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Primary sclerosing cholangitis (PSC) is the classical hepatobiliary manifestation of IBD. This clinical association is linked pathologically to the recruitment of mucosal T cells to the liver, via vascular adhesion protein (VAP)-1-dependent enzyme activity. Our aim was to examine the expression, function and enzymatic activation of the ectoenzyme VAP-1 in patients with PSC. DESIGN We examined VAP-1 expression in patients with PSC, correlated levels with clinical characteristics and determined the functional consequences of enzyme activation by specific enzyme substrates on hepatic endothelium. RESULTS The intrahepatic enzyme activity of VAP-1 was elevated in PSC versus immune-mediated disease controls and non-diseased liver (p<0.001). The adhesion of gut-tropic α4β7+lymphocytes to hepatic endothelial cells in vitro under flow was attenuated by 50% following administration of the VAP-1 inhibitor semicarbazide (p<0.01). Of a number of natural VAP-1 substrates tested, cysteamine-which can be secreted by inflamed colonic epithelium and gut bacteria-was the most efficient (yielded the highest enzymatic rate) and efficacious in its ability to induce expression of functional mucosal addressin cell adhesion molecule-1 on hepatic endothelium. In a prospectively evaluated patient cohort with PSC, elevated serum soluble (s)VAP-1 levels predicted poorer transplant-free survival for patients, independently (HR: 3.85, p=0.003) and additively (HR: 2.02, p=0.012) of the presence of liver cirrhosis. CONCLUSIONS VAP-1 expression is increased in PSC, facilitates adhesion of gut-tropic lymphocytes to liver endothelium in a substrate-dependent manner, and elevated levels of its circulating form predict clinical outcome in patients.
Collapse
Affiliation(s)
- Palak J Trivedi
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
| | - Joseph Tickle
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Mette Nåmdal Vesterhus
- Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway,National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - Peter J Eddowes
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Tony Bruns
- Department of Internal Medicine IV, University Hospital Jena, Germany,Center for Sepsis Control and Care, University Hospital Jena, Germany
| | | | - Richard Parker
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
| | | | - Evaggelia Liaskou
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Liv Wenche Thorbjørnsen
- Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway,National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - Gideon M Hirschfield
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
| | - Kaisa Auvinen
- MediCity Research Laboratory, University of Turku, Turku, Finland,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - Stefan G Hubscher
- Department of Cellular Pathology, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
| | - Marko Salmi
- MediCity Research Laboratory, University of Turku, Turku, Finland,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| | - David H Adams
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK,Liver Unit, University Hospitals Birmingham Queen Elizabeth, Birmingham, UK
| | - Chris J Weston
- National Institute of Health Research Birmingham Liver Biomedical Research Centre Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| |
Collapse
|
50
|
Kusebauch U, Hernández-Castellano LE, Bislev SL, Moritz RL, Røntved CM, Bendixen E. Selected reaction monitoring mass spectrometry of mastitis milk reveals pathogen-specific regulation of bovine host response proteins. J Dairy Sci 2018; 101:6532-6541. [PMID: 29655560 DOI: 10.3168/jds.2017-14312] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/16/2018] [Indexed: 01/29/2023]
Abstract
Mastitis is a major challenge to bovine health. The detection of sensitive markers for mastitis in dairy herds is of great demand. Suitable biomarkers should be measurable in milk and should report pathogen-specific changes at an early stage to support earlier diagnosis and more efficient treatment. However, the identification of sensitive biomarkers in milk has remained a challenge, in part due to their relatively low concentration in milk. In the present study, we used a selected reaction monitoring (SRM) mass spectrometry approach, which allowed the absolute quantitation of 13 host response proteins in milk for the first time. These proteins were measured over a 54-h period upon an in vivo challenge with cell wall components from either gram-negative (lipopolysaccharide from Escherichia coli; LPS) or gram-positive bacteria (peptidoglycan from Staphylococcus aureus; PGN). Whereas our data clearly demonstrate that all challenged animals have consistent upregulation of innate immune response proteins after both LPS and PGN challenge, the data also reveal clearly that LPS challenge unleashes faster and shows a more intense host response compared with PGN challenge. Biomarker candidates that may distinguish between gram-negative and gram-positive bacteria include α-2 macroglobulin, α-1 antitrypsin, haptoglobin, serum amyloid A3, cluster of differentiation 14, calgranulin B, cathepsin C, vanin-1, galectin 1, galectin 3, and IL-8. Our approach can support further studies of large cohorts of animals with natural occurring mastitis, to validate the relevance of these suggested biomarkers in dairy production.
Collapse
Affiliation(s)
| | - Lorenzo E Hernández-Castellano
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; Department of Animal Science, Aarhus University, 8830 Tjele, Denmark
| | - Stine L Bislev
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; Department of Animal Science, Aarhus University, 8830 Tjele, Denmark
| | | | | | - Emøke Bendixen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; Department of Animal Science, Aarhus University, 8830 Tjele, Denmark.
| |
Collapse
|