1
|
Durrington PN, Bashir B, Soran H. How Does HDL Participate in Atherogenesis? Antioxidant Activity Versus Role in Reverse Cholesterol Transport. Antioxidants (Basel) 2025; 14:430. [PMID: 40298833 PMCID: PMC12023944 DOI: 10.3390/antiox14040430] [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: 02/13/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 04/30/2025] Open
Abstract
Low-density lipoprotein (LDL) chemically modified by reactive oxygen species (ROS), for example, leaking from red blood cells in the vascular compartment, more readily crosses the vascular endothelium than does nonoxidatively modified LDL to enter tissue fluid. Oxidatively modified LDL (oxLDL) may also be created in the tissue fluid by ROS leaking from cells by design, for example, by inflammatory white cells, or simply leaking from other cells as a consequence of oxygen metabolism. As well as oxLDL, glycatively modified LDL (glycLDL) is formed in the circulation. High-density lipoprotein (HDL) appears capable of decreasing the burden of lipid peroxides formed on LDL exposed to ROS or to glucose and its metabolites. The mechanism for this that has received the most attention is the antioxidant activity of HDL, which is due in large part to the presence of paraoxonase 1 (PON1). PON1 is intimately associated with its apolipoprotein A1 component and with HDL's lipid domains into which lipid peroxides from LDL or cell membranes can be transferred. It is frequently overlooked that for PON1 to hydrolyze lipid substrates, it is essential that it remain by virtue of its hydrophobic amino acid sequences within a lipid micellar environment, for example, during its isolation from serum or genetically modified cells in tissue culture. Otherwise, it may retain its capacity to hydrolyze water-soluble substrates, such as phenyl acetate, whilst failing to hydrolyze more lipid-soluble molecules. OxLDL and probably glycLDL, once they have crossed the arterial endothelium by receptor-mediated transcytosis, are rapidly taken up by monocytes in a process that also involves scavenger receptors, leading to subendothelial foam cell formation. These are the precursors of atheroma, inducing more monocytes to cross the endothelium into the lesion and the proliferation and migration of myocytes present in the arterial wall into the developing lesion, where they transform into foam cells and fibroblasts. The atheroma progresses to have a central extracellular lake of cholesteryl ester following necrosis and apoptosis of foam cells with an overlying fibrous cap whilst continuing to grow concentrically around the arterial wall by a process involving oxLDL and glycLDL. Within the arterial wall, additional oxLDL is generated by ROS secreted by inflammatory cells and leakage from cells generally when couplet oxygen is reduced. PON1 is important for the mechanism by which HDL opposes atherogenesis, which may provide a better avenue of inquiry in the identification of vulnerable individuals and the provision of new therapies than have emerged from the emphasis placed on its role in RCT.
Collapse
Affiliation(s)
- Paul N. Durrington
- Faculty of Biology, Medicine and Health, Cardiovascular Research Group, University of Manchester, Manchester M13 9NT, UK; (B.B.); (H.S.)
| | - Bilal Bashir
- Faculty of Biology, Medicine and Health, Cardiovascular Research Group, University of Manchester, Manchester M13 9NT, UK; (B.B.); (H.S.)
- Department of Diabetes, Endocrinology and Metabolism, Peter Mount Building, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| | - Handrean Soran
- Faculty of Biology, Medicine and Health, Cardiovascular Research Group, University of Manchester, Manchester M13 9NT, UK; (B.B.); (H.S.)
- Department of Diabetes, Endocrinology and Metabolism, Peter Mount Building, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| |
Collapse
|
2
|
Reichert CO, Levy D, Maselli LMF, da Cunha J, Gualandro SFM, Bydlowski SP. PON-1 and PON-2 Polymorphisms and PON-1 Paraoxonase Activity in People Living with HIV-1. Antioxidants (Basel) 2025; 14:209. [PMID: 40002395 PMCID: PMC11851513 DOI: 10.3390/antiox14020209] [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: 11/08/2024] [Revised: 02/02/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Antiretroviral therapy (ART) has significantly improved the life expectancy of people living with HIV-1 (PLWH). However, prolonged ART use is linked to metabolic alterations and oxidative stress. The paraoxonase (PON) enzymes, especially PON-1 and PON-2, are critical in maintaining antioxidant balance. Their activity can be influenced by polymorphisms such as Q192R and L55M in PON-1 and A148G and S311C in PON-2. This study examines the impact of these polymorphisms on paraoxonase activity, lipid metabolism, and infection markers in PLWH under various ART regimens. This is a case-control study with 525 participants, 175 healthy controls (HC) and 350 PLWH divided into subgroups: T0 (ART-naïve, n = 48), T1 (ART with reverse transcriptase inhibitors, n = 159), and T2 (ART with protease inhibitors, n = 143). Paraoxonase activity was higher in PLWH (123.0; IQR: 62.0-168.0) compared to HC (91.0; IQR: 48.0-136.0, p < 0.001) but similar between HC and T0 (p = 0.594). T1 (125.0; IQR: 65.5-166.0) and T2 (123.0; IQR: 61.0-182.0) showed higher activity than HC (p = 0.002 and 0.003). Among 61 complete genotypes, 13 were unique to PLWH and 6 to HC (p < 0.001). L55L was more frequent in HC (49.7% vs. 36.9% in PLWH), while M55M was higher in PLWH (p = 0.004). The S311C genotype was more frequent in HC (39.2%) than PLWH (24.9%) (p = 0.003). The L55L genotype conferred 59.9% protection against HIV-1 (OR: 0.401; 95% CI: 0.228-0.704), while the M allele increased susceptibility by ~69% (OR: 1.694; 95% CI: 1.173-2.446). The M55M genotype and/or M allele may be linked to HIV-1 susceptibility. Prolonged ART use elevates PON-1 activity in PLWH.
Collapse
Affiliation(s)
- Cadiele Oliana Reichert
- Lipids, Oxidation, and Cell Biology Group, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil or (C.O.R.); (D.L.); (L.M.F.M.); (J.d.C.)
| | - Débora Levy
- Lipids, Oxidation, and Cell Biology Group, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil or (C.O.R.); (D.L.); (L.M.F.M.); (J.d.C.)
| | - Luciana Morganti Ferreira Maselli
- Lipids, Oxidation, and Cell Biology Group, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil or (C.O.R.); (D.L.); (L.M.F.M.); (J.d.C.)
| | - Joel da Cunha
- Lipids, Oxidation, and Cell Biology Group, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil or (C.O.R.); (D.L.); (L.M.F.M.); (J.d.C.)
| | - Sandra Fátima Menosi Gualandro
- Department of Hematology, Hemotherapy, and Cell Therapy, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo 05419-000, SP, Brazil;
| | - Sérgio Paulo Bydlowski
- Lipids, Oxidation, and Cell Biology Group, Laboratory of Immunology (LIM19), Heart Institute (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil or (C.O.R.); (D.L.); (L.M.F.M.); (J.d.C.)
- Department of Hematology, Hemotherapy, and Cell Therapy, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo 05419-000, SP, Brazil;
- Instituto Nacional de Ciencia e Tecnologia em Medicina Regenerativa (INCT-Regenera), CNPq, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
3
|
Zargari M, Maadi N, Rezapour M, Bagheri A, Fallahpour S, Nosrati M, Mahrooz A. The Regulatory Variant -108C/T in the Promoter of Paraoxonase 1 (PON1) Gene has a More Important Role in Regulating PON1 Activity Compared to rs3735590 in 3'-UTR in Patients with Coronary Artery Disease. Adv Biomed Res 2024; 13:38. [PMID: 39224397 PMCID: PMC11368222 DOI: 10.4103/abr.abr_391_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/20/2024] [Accepted: 03/02/2024] [Indexed: 09/04/2024] Open
Abstract
Background This study aimed to assess the serum activity of paraoxonase 1 (PON1) in patients with coronary artery disease (CAD) based on two genetic variants including the -108C/T variant in the promoter region and the rs3735590 variant in the binding site of miR-616 at the 3'-UTR of the PON1 gene. Materials and Methods A total of 140 subjects who exhibited clinical symptoms of CAD underwent diagnostic coronary angiography. The patients with CAD were further categorized into two groups: single-vessel disease (SVD) and multi-vessel disease (MVD). The study variants were genotyped using the restriction fragment length polymorphism (RFLP) technique after polymerase chain reaction amplification. Results After adjusting for age, gender, body mass index, metformin, and statin usage, a significant association was observed between the -108C/T variant and PON1 activity (P < 0.001). In the sub-groups of both SVD and MVD, individuals with the TC+CC genotypes exhibited significantly higher PON1 activity compared to TT homozygotes (P = 0.001 for SVD and P = 0.01 for MVD). As for the rs3735590 variant, individuals with the A allele (GA+AA genotypes) had higher PON1 activity compared to those with the GG genotype in both the SVD and MVD groups, although the results did not reach statistical significance. Conclusions Our study findings indicate a significant decrease in PON1 activity among patients with obstructive CAD. Notably, our results suggest that the -108C/T variant exerts a greater influence on PON1 activity compared to the rs3735590 variant. These findings highlight the crucial role of the -108C/T variant in modulating PON1 activity within the context of atherosclerosis.
Collapse
Affiliation(s)
- Mehryar Zargari
- Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Negar Maadi
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maysam Rezapour
- Department of Paramedicine, Amol Paramedical Sciences School, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abouzar Bagheri
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Samane Fallahpour
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mani Nosrati
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdolkarim Mahrooz
- Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
4
|
Sirca TB, Mureșan ME, Pallag A, Marian E, Jurca T, Vicaș LG, Tunduc IP, Manole F, Ștefan L. The Role of Polyphenols in Modulating PON1 Activity Regarding Endothelial Dysfunction and Atherosclerosis. Int J Mol Sci 2024; 25:2962. [PMID: 38474211 DOI: 10.3390/ijms25052962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/24/2024] [Accepted: 03/02/2024] [Indexed: 03/14/2024] Open
Abstract
The incidence and prevalence of cardiovascular diseases are still rising. The principal mechanism that drives them is atherosclerosis, an affection given by dyslipidemia and a pro-inflammatory state. Paraoxonase enzymes have a protective role due to their ability to contribute to antioxidant and anti-inflammatory pathways, especially paraoxonase 1 (PON1). PON1 binds with HDL (high-density lipoprotein), and high serum levels lead to a protective state against dyslipidemia, cardiovascular diseases, diabetes, stroke, nonalcoholic fatty liver disease, and many others. Modulating PON1 expression might be a treatment objective with significant results in limiting the prevalence of atherosclerosis. Lifestyle including diet and exercise can raise its levels, and some beneficial plants have been found to influence PON1 levels; therefore, more studies on herbal components are needed. Our purpose is to highlight the principal roles of Praoxonase 1, its implications in dyslipidemia, cardiovascular diseases, stroke, and other diseases, and to emphasize plants that can modulate PON1 expression, targeting the potential of some flavonoids that could be introduced as supplements in our diet and to validate the hypothesis that flavonoids have any effects regarding PON1 function.
Collapse
Affiliation(s)
- Teodora Bianca Sirca
- Doctoral School of Biomedical Sciences, University of Oradea, No. 1 University Street, 410087 Oradea, Romania
| | - Mariana Eugenia Mureșan
- Doctoral School of Biomedical Sciences, University of Oradea, No. 1 University Street, 410087 Oradea, Romania
| | - Annamaria Pallag
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania
| | - Eleonora Marian
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania
| | - Tunde Jurca
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania
| | - Laura Grațiela Vicaș
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 29 Nicolae Jiga Street, 410028 Oradea, Romania
| | - Ioana Paula Tunduc
- Department of Cardiology, Clinical County Emergency Hospital of Bihor, Gheorghe Doja Street 65-67, 410169 Oradea, Romania
| | - Felicia Manole
- Department of Surgery, Faculty of Medicine and Pharmacy, University of Oradea, 1st December Square 10, 410073 Oradea, Romania
| | - Liana Ștefan
- Department of Surgery, Faculty of Medicine and Pharmacy, University of Oradea, 1st December Square 10, 410073 Oradea, Romania
| |
Collapse
|
5
|
Mahrooz A, Khosravi-Asrami OF, Alizadeh A, Mohmmadi N, Bagheri A, Kashi Z, Bahar A, Nosrati M, Mackness M. Can HDL cholesterol be replaced by paraoxonase 1 activity in the prediction of severe coronary artery disease in patients with type 2 diabetes? Nutr Metab Cardiovasc Dis 2023; 33:1599-1607. [PMID: 37344284 DOI: 10.1016/j.numecd.2023.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 04/06/2023] [Accepted: 05/12/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND AND AIMS Novel biomarkers are required to improve cardiovascular disease prediction in patients with type 2 diabetes (T2D) as a high-risk population. This study was conducted to examine whether coronary artery disease (CAD) risk assessment can be improved by substituting high-density lipoprotein (HDL)-bound paraoxonase 1 (PON1) activity for HDL cholesterol (HDL-C) concentration in patients with T2D. METHODS AND RESULTS In this study, we studied 139 patients with T2D (mean age 64.12 ± 8.17 years) who underwent coronary angiographic examination. The initial rate of substrate hydrolysis was spectrophotometrically assayed in kinetic mode for measuring PON1 activity. Receiver operating characteristic (ROC) graphs are created by plotting true positivity versus false positivity. In patients with HbA1c ≥ 7%, PON1 (AUC = 0.7, p = 0.029) and nonHDL-C/PON1 (AUC = 0.75, p = 0.013) were significantly more capable of differentiating patients with CAD from those without CAD compared to HDL-C and nonHDL-C/HDL-C. Also, the predictive power of PON1 (AUC = 0.64, p = 0.029) and nonHDL-C/PON1 (AUC = 0.71, p = 0.004) were significantly higher in comparison with HDL-C and nonHDL-C/HDL-C for CAD characterization in patients aged ≥50 years. Moreover, PON1 and nonHDL-C/PON1 are associated with the incidence of CAD with an AUC of 0.7 (p = 0.026) and AUC of 0.64 (p = 0.087), respectively, among subjects with low HDL-C. CONCLUSION PON1 and the ratio of nonHDL-C/PON1 significantly improve the prediction of severe CAD in T2D patients and in patients with HbA1c ≥ 7%, age ≥50 years, or low HDL-C. PON1 activity and lipid ratios using this enzyme may be valuable as substitutes of HDL-C for increasing clinical efficacies in cardiovascular risk assessment.
Collapse
Affiliation(s)
- Abdolkarim Mahrooz
- Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Diabetes Research Center, Imam Teaching Hospital, Mazandaran University of Medical Sciences, Sari, Iran; Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Omeh Farveh Khosravi-Asrami
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ahad Alizadeh
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Neda Mohmmadi
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Abouzar Bagheri
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Kashi
- Diabetes Research Center, Imam Teaching Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Adele Bahar
- Diabetes Research Center, Imam Teaching Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mani Nosrati
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mike Mackness
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
6
|
Kunachowicz D, Ściskalska M, Kepinska M. Modulatory Effect of Lifestyle-Related, Environmental and Genetic Factors on Paraoxonase-1 Activity: A Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2813. [PMID: 36833509 PMCID: PMC9957543 DOI: 10.3390/ijerph20042813] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Paraoxonase-1 (PON1) is a calcium-dependent, HDL-bound serum hydrolase active toward a wide variety of substrates. PON1 displays three types of activities, among which lactonase, paraoxonase, arylesterase and phosphotriesterase can be distinguished. Not only is this enzyme a major organophosphate compound detoxifier, but it is also an important constituent of the cellular antioxidant system and has anti-inflammatory and antiatherogenic functions. The concentration and activity of PON1 is highly variable among individuals, and these differences can be both of genetic origin and be a subject of epigenetic regulation. Owing to the fact that, in recent decades, the exposure of humans to an increasing number of different xenobiotics has been continuously rising, the issues concerning the role and activity of PON1 shall be reconsidered with particular attention to growing pharmaceuticals intake, dietary habits and environmental awareness. In the following manuscript, the current state of knowledge concerning the influence of certain modifiable and unmodifiable factors, including smoking, alcohol intake, gender, age and genotype variation on PON1 activity, along with pathways through which these could interfere with the enzyme's protective functions, is presented and discussed. Since exposure to certain xenobiotics plays a key role in PON1 activity, the influence of organophosphates, heavy metals and several pharmaceutical agents is also specified.
Collapse
Affiliation(s)
| | | | - Marta Kepinska
- Department of Pharmaceutical Biochemistry, Division of Biomedical and Environmental Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50–556 Wrocław, Poland
| |
Collapse
|
7
|
Okamoto J, Wang L, Yin X, Luca F, Pique-Regi R, Helms A, Im HK, Morrison J, Wen X. Probabilistic integration of transcriptome-wide association studies and colocalization analysis identifies key molecular pathways of complex traits. Am J Hum Genet 2023; 110:44-57. [PMID: 36608684 PMCID: PMC9892769 DOI: 10.1016/j.ajhg.2022.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/06/2022] [Indexed: 01/07/2023] Open
Abstract
Integrative genetic association methods have shown great promise in post-GWAS (genome-wide association study) analyses, in which one of the most challenging tasks is identifying putative causal genes and uncovering molecular mechanisms of complex traits. Recent studies suggest that prevailing computational approaches, including transcriptome-wide association studies (TWASs) and colocalization analysis, are individually imperfect, but their joint usage can yield robust and powerful inference results. This paper presents INTACT, a computational framework to integrate probabilistic evidence from these distinct types of analyses and implicate putative causal genes. This procedure is flexible and can work with a wide range of existing integrative analysis approaches. It has the unique ability to quantify the uncertainty of implicated genes, enabling rigorous control of false-positive discoveries. Taking advantage of this highly desirable feature, we further propose an efficient algorithm, INTACT-GSE, for gene set enrichment analysis based on the integrated probabilistic evidence. We examine the proposed computational methods and illustrate their improved performance over the existing approaches through simulation studies. We apply the proposed methods to analyze the multi-tissue eQTL data from the GTEx project and eight large-scale complex- and molecular-trait GWAS datasets from multiple consortia and the UK Biobank. Overall, we find that the proposed methods markedly improve the existing putative gene implication methods and are particularly advantageous in evaluating and identifying key gene sets and biological pathways underlying complex traits.
Collapse
Affiliation(s)
- Jeffrey Okamoto
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Lijia Wang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xianyong Yin
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francesca Luca
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Adam Helms
- University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Hae Kyung Im
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jean Morrison
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiaoquan Wen
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
8
|
Hasheminasabgorji E, Mishan MA, Tabari MAK, Bagheri A. miR-638: A Promising Cancer Biomarker with Therapeutic Potential. Curr Mol Med 2023; 23:377-389. [PMID: 35382724 DOI: 10.2174/1566524022666220405125900] [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: 10/26/2021] [Revised: 02/03/2022] [Accepted: 02/16/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND There is an unmet need to improve the diagnosis of cancer with precise treatment strategies. Therefore, more powerful diagnostic, prognostic, and therapeutic biomarkers are needed to overcome tumor cells. microRNAs (miRNAs, miRs), as a class of small non-coding RNAs, play essential roles in cancer through the tumor-suppressive or oncogenic effects by post-transcriptional regulation of their targets. Many studies have provided shreds of evidence on aberrantly expressed miRNAs in numerous cancers and have shown that miRNAs could play potential roles as diagnostic, prognostic, and even therapeutic biomarkers in patients with cancers. Findings have revealed that miR-638 over or underexpression might play a critical role in cancer initiation, development, and progression. However, the mechanistic effects of miR-638 on cancer cells are still controversial. CONCLUSION In the present review, we have focused on the diagnostic, prognostic, and therapeutic potentials of miR-638 and discussed its mechanistic roles in various types of cancers.
Collapse
Affiliation(s)
- Elham Hasheminasabgorji
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Arnie Charbonneau Cancer Institute, Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mohammad Amir Mishan
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
- USERN Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abouzar Bagheri
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
9
|
Istratoaie S, Boroş B, Vesa ŞC, Maria Pop R, Cismaru G, Pop D, Vasile Milaciu M, Ciumărnean L, Văcăraş V, Dana Buzoianu A. Paraoxonase 1 and atrial fibrillation: Is there a relationship? Medicine (Baltimore) 2022; 101:e31553. [PMID: 36401436 PMCID: PMC9678507 DOI: 10.1097/md.0000000000031553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/05/2022] [Indexed: 12/03/2022] Open
Abstract
Atrial fibrillation (AF) is associated with oxidative stress and inflammation. Paraoxonase-1 (PON1), circulates in blood bound to high-density lipoproteins and reduces systemic oxidative stress. The aim of this study was to evaluate PON1 serum concentration and PON1 arylesterase activity (AREase) in patients with AF. We studied a group of 67 patients with symptomatic paroxysmal or persistent AF admitted for cardioversion and a control group of 59 patients without AF. Clinical parameters, lipid profile, PON1 concentration and AREase were evaluated. A significant difference in serum PON1 concentration and in AREase was found among the two groups. In a multivariate linear regression model, the presence of AF was associated with low PON1 concentration (P = .022). The body mass index was also independently associated with PON1 values (P < .001). Only the high-density lipoproteins-cholesterol level was independently associated with AREase (P = .002). PON1 serum concentrations and AREase were diminished in patients with AF, and the presence of AF was independently associated with low PON1 values.
Collapse
Affiliation(s)
- Sabina Istratoaie
- Department of Pharmacology, Toxicology and Clinical Pharmacology, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Bianca Boroş
- 5th Department of Internal Medicine, Cardiology-Rehabilitation, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ştefan Cristian Vesa
- Department of Pharmacology, Toxicology and Clinical Pharmacology, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Raluca Maria Pop
- Department of Pharmacology, Toxicology and Clinical Pharmacology, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriel Cismaru
- 5th Department of Internal Medicine, Cardiology-Rehabilitation, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Dana Pop
- 5th Department of Internal Medicine, Cardiology-Rehabilitation, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mircea Vasile Milaciu
- 5th Department of Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lorena Ciumărnean
- 5th Department of Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Vitalie Văcăraş
- Department of Neurosciences, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Anca Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
10
|
Soflaei SS, Baktashian M, Moghaddam KH, Saberi-Karimian M, Kosari N, Hashemi SM, Mouhebati M, Amini M, Dehghani M, Esmaily H, Ebrahimi M, Falsoleiman H, Nosrati-Tirkani A, Sadabadi F, Ferns GA, Salehi M, Pasdar A, Ghayour-Mobarhan M. Association of Paraoxonase-1 Genotype and Phenotype with Angiogram Positive Coronary Artery Disease. Arq Bras Cardiol 2022; 119:593-601. [PMID: 36074479 PMCID: PMC9563872 DOI: 10.36660/abc.20210422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 03/02/2022] [Accepted: 04/06/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND It has been shown that increased serum PON1 levels are protective against several disorders. Several single nucleotide polymorphisms (SNPs) of the PON1 gene have been reported to be associated with serum enzyme protein levels and activity. OBJECTIVE To investigate the association of SNPs of PON1 and serum paraoxonase activity with coronary artery disease (CAD). METHODS A total of 601 unrelated patients who underwent coronary angiography including those who had >50% stenosis (N=266) and those with <30% stenosis (N=335) were studied. The Paraoxonase gene rs662 and rs840560 SNPs were determined using the ARMS-PCR method and the rs705379 SNP was genotyped using PCR-RFLP analysis. Serum paraoxonase activity was measured using paraoxon as a substrate. A p value of p<0.05 was considered as significant. RESULTS Serum paraoxonase activity was not significantly different between the study groups. After adjustment for age, sex, hypertension, diabetes mellitus and dyslipidemia, the GG genotype and co-dominant model of rs662 was positively associated with a positive angiogram (respectively, OR=2.424, 95%CI [1.123-5.233], p<0.05, OR=1.663, 95%CI [1.086-2.547]). Serum paraoxonase activity was significantly higher in the G allele and GG variant of rs662, A allele and AA variant of rs854560 and C allele and CC variant of rs705379. The haplotype analysis has shown that the ATC haplotype was significantly more prevalent among the angiogram negative group. The analysis between groups indicated that the A allele of rs662 was significantly associated with lower paraoxonase activity in the positive angiogram group (p=0.019). CONCLUSIONS The presence of the G allele of the rs662 single nucleotide polymorphism is independently associated to increased risk of CAD.
Collapse
Affiliation(s)
- Sara Saffar Soflaei
- International UNESCO Center for Health-Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIrã International UNESCO Center for Health-Related Basic Sciences and Human Nutrition , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Mojtaba Baktashian
- International UNESCO Center for Health-Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIrã International UNESCO Center for Health-Related Basic Sciences and Human Nutrition , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Kiana Hosseinpour Moghaddam
- Department of BiologyFaculty of SciencesFerdowsi University of MashhadMashhadIrã Department of Biology , Faculty of Sciences , Ferdowsi University of Mashhad , Mashhad – Irã
| | - Maryam Saberi-Karimian
- Metabolic Syndrome Research CenterFaculty of MedicineMashhad University of Medical SciencesMashhadIrã Metabolic Syndrome Research Center , Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Negin Kosari
- International UNESCO Center for Health-Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIrã International UNESCO Center for Health-Related Basic Sciences and Human Nutrition , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Seyed Mohammad Hashemi
- Department of CardiologySchool of MedicineIsfahan University of Medical SciencesIsfahanIrã Department of Cardiology , Chamran Hospital, School of Medicine , Isfahan University of Medical Sciences , Isfahan – Irã
| | - Mohsen Mouhebati
- Cardiovascular Research CenterSchool of MedicineMashhad University of Medical SciencesMashhadIrã Cardiovascular Research Center , School of Medicine , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Mahsa Amini
- International UNESCO Center for Health-Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIrã International UNESCO Center for Health-Related Basic Sciences and Human Nutrition , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Mashallah Dehghani
- Cardiovascular Research CenterSchool of MedicineMashhad University of Medical SciencesMashhadIrã Cardiovascular Research Center , School of Medicine , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Habibollah Esmaily
- Department of Biostatistics and EpidemiologySchool of HealthMashhad University of Medical SciencesMashhadIrã Department of Biostatistics and Epidemiology , School of Health , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Mahmoud Ebrahimi
- Cardiovascular Research CenterSchool of MedicineMashhad University of Medical SciencesMashhadIrã Cardiovascular Research Center , School of Medicine , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Homa Falsoleiman
- Cardiovascular Research CenterSchool of MedicineMashhad University of Medical SciencesMashhadIrã Cardiovascular Research Center , School of Medicine , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Abolfazl Nosrati-Tirkani
- Biochemistry of Nutrition Research CenterFaculty of MedicineMashhad University of Medical SciencesMashhadIrã Biochemistry of Nutrition Research Center , Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Fatemeh Sadabadi
- International UNESCO Center for Health-Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIrã International UNESCO Center for Health-Related Basic Sciences and Human Nutrition , Mashhad University of Medical Sciences , Mashhad – Irã
| | - Gordon A. Ferns
- Brighton & Sussex Medical SchoolDivision of Medical EducationBrightonReino Unido Brighton & Sussex Medical School , Division of Medical Education , Falmer, Brighton – Reino Unido
| | - Mansoor Salehi
- Department of geneticsFaculty of medicine and genetics laboratory AL Zahra hospitalIsfahan University of MedicineIsfahanIrã Department of genetics , Faculty of medicine and genetics laboratory AL Zahra hospital , Isfahan University of Medicine , Isfahan – Irã
| | - Alireza Pasdar
- International UNESCO Center for Health-Related Basic Sciences and Human NutritionMashhad University of Medical SciencesMashhadIrã International UNESCO Center for Health-Related Basic Sciences and Human Nutrition , Mashhad University of Medical Sciences , Mashhad – Irã
- Division of Applied MedicineMedical SchoolUniversity of AberdeenAberdeenReino Unido Division of Applied Medicine , Medical School , University of Aberdeen , Foresterhill, Aberdeen – Reino Unido
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research CenterFaculty of MedicineMashhad University of Medical SciencesMashhadIrã Metabolic Syndrome Research Center , Faculty of Medicine , Mashhad University of Medical Sciences , Mashhad – Irã
- Cardiovascular Research CenterSchool of MedicineMashhad University of Medical SciencesMashhadIrã Cardiovascular Research Center , School of Medicine , Mashhad University of Medical Sciences , Mashhad – Irã
| |
Collapse
|
11
|
Otocka-Kmiecik A. Effect of Carotenoids on Paraoxonase-1 Activity and Gene Expression. Nutrients 2022; 14:nu14142842. [PMID: 35889799 PMCID: PMC9318174 DOI: 10.3390/nu14142842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 12/27/2022] Open
Abstract
Paraoxonase 1 (PON1) is an antioxidant enzyme attached to HDL with an anti-atherogenic potential. It protects LDL and HDL from lipid peroxidation. The enzyme is sensitive to various modulating factors, such as genetic polymorphisms as well as pharmacological, dietary (including carotenoids), and lifestyle interventions. Carotenoids are nutritional pigments with antioxidant activity. The aim of this review was to gather evidence on their effect on the modulation of PON1 activity and gene expression. Carotenoids administered as naturally occurring nutritional mixtures may present a synergistic beneficial effect on PON1 status. The effect of carotenoids on the enzyme depends on age, ethnicity, gender, diet, and PON1 genetic variation. Carotenoids, especially astaxanthin, β-carotene, and lycopene, increase PON1 activity. This effect may be explained by their ability to quench singlet oxygen and scavenge free radicals. β-carotene and lycopene were additionally shown to upregulate PON1 gene expression. The putative mechanisms of such regulation involve PON1 CpG-rich region methylation, Ca(2+)/calmodulin-dependent kinase II (CaMKKII) pathway induction, and upregulation via steroid regulatory element-binding protein-2 (SREBP-2). More detailed and extensive research on the mechanisms of PON1 modulation by carotenoids may lead to the development of new targeted therapies for cardiovascular diseases.
Collapse
Affiliation(s)
- Aneta Otocka-Kmiecik
- Department of Experimental Physiology, Medical University of Lodz, 6/8 Mazowiecka St., 92-215 Lodz, Poland
| |
Collapse
|
12
|
Arab ZN, Khayatan D, Razavi SM, Zare K, Kheradkhah E, Momtaz S, Ferretti G, Bacchetti T, Sathyapalan T, Emami SA, Abdolghaffari AH, Sahebkar A. Phytochemicals as Modulators of Paraoxonase-1 in Health and Diseases. Antioxidants (Basel) 2022; 11:1273. [PMID: 35883764 PMCID: PMC9311866 DOI: 10.3390/antiox11071273] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 02/05/2023] Open
Abstract
Chronic diseases such as cardiovascular disease (CVD), atherosclerosis, chronic liver disease, and neurodegenerative diseases are major causes of mortality. These diseases have gained much attention due to their complications, and therefore novel approaches with fewer side effects are an important research topic. Free radicals and oxidative stress are involved in the molecular mechanisms of several diseases. Antioxidants can scavenge free radicals and mitigate their adverse effects. One of the most important antioxidant enzymes are paraoxonases (PONs). These enzymes perform a wide range of physiological activities ranging from drug metabolism to detoxification of neuroleptics. Paraoxonase-1 (PON1) is produced in the liver and then transferred to the bloodstream. It has been demonstrated that PON1 could have beneficial effects in numerous diseases such as atherosclerosis, CVD, diabetes mellitus, and neurodegenerative diseases by modulating relevant signalling pathways involved in inflammation and oxidative stress. These pathways include peroxisome proliferator-activated receptor gamma (PPAR-γ) and protein kinase B/nuclear factor kappa-light-chain-enhancer of activated B cells (AKT/NF-κB)-dependent signalling pathways. Increasing PON1 could potentially have protective effects and reduce the incidence of various diseases by modulating these signalling pathways. Several studies have reported that dietary factors are able to modulate PON1 expression and activity. This review aimed at summarizing the state of the art on the effects of dietary phytochemicals on PON1 enzyme activity and the relevant signalling pathways in different diseases.
Collapse
Affiliation(s)
- Zahra Najafi Arab
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; (Z.N.A.); (D.K.); (S.M.R.); (E.K.)
| | - Danial Khayatan
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; (Z.N.A.); (D.K.); (S.M.R.); (E.K.)
| | - Seyed Mehrad Razavi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; (Z.N.A.); (D.K.); (S.M.R.); (E.K.)
| | - Kimia Zare
- School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran;
| | - Elnaz Kheradkhah
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; (Z.N.A.); (D.K.); (S.M.R.); (E.K.)
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran;
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), the Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Gianna Ferretti
- Department of Clinical Science, Polytechnic University of Marche, Ancona, Italy
| | - Tiziana Bacchetti
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy;
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull HU3 2JZ, UK;
| | - Seyed Ahmad Emami
- Department of Traditional Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran;
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; (Z.N.A.); (D.K.); (S.M.R.); (E.K.)
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran;
- Department of Toxicology and Pharmacology, School of Pharmacy, and Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), the Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Paraoxonase-1 Facilitates PRRSV Replication by Interacting with Viral Nonstructural Protein-9 and Inhibiting Type I Interferon Pathway. Viruses 2022; 14:v14061203. [PMID: 35746674 PMCID: PMC9230610 DOI: 10.3390/v14061203] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/29/2022] [Accepted: 05/29/2022] [Indexed: 02/04/2023] Open
Abstract
Paraoxonase-1 (PON1), an esterase with specifically paraoxonase activity, has been proven to be involved in inflammation and infection. Porcine reproductive and respiratory syndrome virus (PRRSV) is still a major concern in pigs and causes severe economic losses to the swine industry worldwide. In this study, the role of PON1 was investigated in porcine alveolar macrophages (PAMs) during PRRSV infection. The results showed that PRRSV replication downregulated PON1, and the knockdown of PON1 significantly decreased PRRSV replication. Similarly, PON1 overexpression could enhance PRRSV replication. Interestingly, we observed that PON1 interacted with PRRSV nonstructural protein 9 (Nsp9), the RNA-dependent RNA polymerase, and the knockdown of PON1 lowered the RNA binding ability of Nsp9, suggesting that PON1 can facilitate Nsp9 function in viral replication. In addition, the knockdown of PON1 expression led to the amplification of type I interferon (IFN) genes and vice versa. In summary, our data demonstrate that PON1 facilitates PRRSV replication by interacting with Nsp9 and inhibiting the type I IFN signaling pathway. Hence, PON1 may be an additional component of the anti-PRRSV defenses.
Collapse
|
14
|
Chen YP, Wang KX, Cai JQ, Li Y, Yu HL, Wu Q, Meng W, Wang H, Yin CH, Wu J, Huang MB, Li R, Guan DG. Detecting Key Functional Components Group and Speculating the Potential Mechanism of Xiao-Xu-Ming Decoction in Treating Stroke. Front Cell Dev Biol 2022; 10:753425. [PMID: 35646921 PMCID: PMC9136080 DOI: 10.3389/fcell.2022.753425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
Stroke is a cerebrovascular event with cerebral blood flow interruption which is caused by occlusion or bursting of cerebral vessels. At present, the main methods in treating stroke are surgical treatment, statins, and recombinant tissue-type plasminogen activator (rt-PA). Relatively, traditional Chinese medicine (TCM) has widely been used at clinical level in China and some countries in Asia. Xiao-Xu-Ming decoction (XXMD) is a classical and widely used prescription in treating stroke in China. However, the material basis of effect and the action principle of XXMD are still not clear. To solve this issue, we designed a new system pharmacology strategy that combined targets of XXMD and the pathogenetic genes of stroke to construct a functional response space (FRS). The effective proteins from this space were determined by using a novel node importance calculation method, and then the key functional components group (KFCG) that could mediate the effective proteins was selected based on the dynamic programming strategy. The results showed that enriched pathways of effective proteins selected from FRS could cover 99.10% of enriched pathways of reference targets, which were defined by overlapping of component targets and pathogenetic genes. Targets of optimized KFCG with 56 components can be enriched into 166 pathways that covered 80.43% of 138 pathways of 1,012 pathogenetic genes. A component potential effect score (PES) calculation model was constructed to calculate the comprehensive effective score of components in the components-targets-pathways (C-T-P) network of KFCGs, and showed that ferulic acid, zingerone, and vanillic acid had the highest PESs. Prediction and docking simulations show that these components can affect stroke synergistically through genes such as MEK, NFκB, and PI3K in PI3K-Akt, cAMP, and MAPK cascade signals. Finally, ferulic acid, zingerone, and vanillic acid were tested to be protective for PC12 cells and HT22 cells in increasing cell viabilities after oxygen and glucose deprivation (OGD). Our proposed strategy could improve the accuracy on decoding KFCGs of XXMD and provide a methodologic reference for the optimization, mechanism analysis, and secondary development of the formula in TCM.
Collapse
Affiliation(s)
- Yu-peng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Ke-xin Wang
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, National Key Clinical Specialty/Engineering Technology Research Center of Education Ministry of China, Neurosurgery Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jie-qi Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Yi Li
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hai-lang Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Qi Wu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Handuo Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Chuan-hui Yin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Jie Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Mian-bo Huang
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,*Correspondence: Mian-bo Huang, ; Rong Li, ; Dao-gang Guan,
| | - Rong Li
- Department of Cardiovascular Disease, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Mian-bo Huang, ; Rong Li, ; Dao-gang Guan,
| | - Dao-gang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China,*Correspondence: Mian-bo Huang, ; Rong Li, ; Dao-gang Guan,
| |
Collapse
|
15
|
Medina-Díaz IM, Ponce-Ruíz N, Rojas-García AE, Zambrano-Zargoza JF, Bernal-Hernández YY, González-Arias CA, Barrón-Vivanco BS, Herrera-Moreno JF. The Relationship between Cancer and Paraoxonase 1. Antioxidants (Basel) 2022; 11:antiox11040697. [PMID: 35453382 PMCID: PMC9028432 DOI: 10.3390/antiox11040697] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/13/2022] Open
Abstract
Extensive research has been carried out to understand and elucidate the mechanisms of paraoxonase 1 (PON1) in the development of diseases including cancer, cardiovascular diseases, neurological diseases, and inflammatory diseases. This review focuses on the relationship between PON1 and cancer. The data suggest that PON1, oxidative stress, chronic inflammation, and cancer are closely linked. Certainly, the gene expression of PON1 will remain challenging to study. Therefore, targeting PON1, redox-sensitive pathways, and transcription factors promise prevention and therapy in the development of several diseases, including cancer.
Collapse
Affiliation(s)
- Irma Martha Medina-Díaz
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Tepict 63000, Mexico; (N.P.-R.); (A.E.R.-G.); (Y.Y.B.-H.); (C.A.G.-A.); (B.S.B.-V.); (J.F.H.-M.)
- Correspondence:
| | - Néstor Ponce-Ruíz
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Tepict 63000, Mexico; (N.P.-R.); (A.E.R.-G.); (Y.Y.B.-H.); (C.A.G.-A.); (B.S.B.-V.); (J.F.H.-M.)
| | - Aurora Elizabeth Rojas-García
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Tepict 63000, Mexico; (N.P.-R.); (A.E.R.-G.); (Y.Y.B.-H.); (C.A.G.-A.); (B.S.B.-V.); (J.F.H.-M.)
| | | | - Yael Y. Bernal-Hernández
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Tepict 63000, Mexico; (N.P.-R.); (A.E.R.-G.); (Y.Y.B.-H.); (C.A.G.-A.); (B.S.B.-V.); (J.F.H.-M.)
| | - Cyndia Azucena González-Arias
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Tepict 63000, Mexico; (N.P.-R.); (A.E.R.-G.); (Y.Y.B.-H.); (C.A.G.-A.); (B.S.B.-V.); (J.F.H.-M.)
| | - Briscia S. Barrón-Vivanco
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Tepict 63000, Mexico; (N.P.-R.); (A.E.R.-G.); (Y.Y.B.-H.); (C.A.G.-A.); (B.S.B.-V.); (J.F.H.-M.)
| | - José Francisco Herrera-Moreno
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Tepict 63000, Mexico; (N.P.-R.); (A.E.R.-G.); (Y.Y.B.-H.); (C.A.G.-A.); (B.S.B.-V.); (J.F.H.-M.)
| |
Collapse
|
16
|
Abstract
Paraoxonase 2 (PON2) is a ubiquitously expressed intracellular enzyme that is known to have a protective role from oxidative stress. Clinical studies have also demonstrated the significance of PON2 in the manifestation of cardiovascular and several other diseases, and hence, it is considered an important biomarker. Recent findings of its expression in brain tissue suggest its potential protective effect on oxidative stress and neuroinflammation. Polymorphisms of PON2 in humans are a risk factor in many pathological conditions, suggesting a possible mechanism of its anti-oxidative property probably through lactonase activity. However, exogenous factors may also modulate the expression and activity of PON2. Hence, this review aims to report the mechanism by which PON2 expression is regulated and its role in oxidative stress disorders such as neurodegeneration and tumor formation. The role of PON2 owing to its lactonase activity in bacterial infectious diseases and association of PON2 polymorphism with pathological conditions are also highlighted.
Collapse
Affiliation(s)
- Fauzia Parween
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| | - Rinkoo Devi Gupta
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
| |
Collapse
|
17
|
Khazeei Tabari MA, Mishan MA, Moradi M, Khandan M, Khoshhal H, Mahrooz A, Bagheri A. Noncoding RNA Roles in Pharmacogenomic Responses to Aspirin: New Molecular Mechanisms for an Old Drug. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6830560. [PMID: 34926688 PMCID: PMC8677408 DOI: 10.1155/2021/6830560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022]
Abstract
Aspirin, as one of the most frequently prescribed drugs, can have therapeutic effects on different conditions such as cardiovascular and metabolic disorders and malignancies. The effects of this common cardiovascular drug are exerted through different molecular and cellular pathways. Altered noncoding RNA (ncRNA) expression profiles during aspirin treatments indicate a close relationship between these regulatory molecules and aspirin effects through regulating gene expressions. A better understanding of the molecular networks contributing to aspirin efficacy would help optimize efficient therapies for this very popular drug. This review is aimed at discussing and highlighting the identified interactions between aspirin and ncRNAs and their targeting pathways and better understanding pharmacogenetic responses to aspirin.
Collapse
Affiliation(s)
- Mohammad Amin Khazeei Tabari
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
- USERN Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Amir Mishan
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Moradi
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohanna Khandan
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
- USERN Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hooman Khoshhal
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
- USERN Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdolkarim Mahrooz
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abouzar Bagheri
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Molecular and Cell Biology Research Center, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Clinical Biochemistry and Medical Genetics, Gastrointestinal Cancer Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
18
|
Pires RS, Braga PGS, Santos JMB, Amaral JB, Amirato GR, Trettel CS, Dos Santos CAF, Vaisberg M, Nali LHS, Vieira RP, Maranhão RC, Pithon-Curi TC, Barros MP, Bachi ALL. l-Glutamine supplementation enhances glutathione peroxidase and paraoxonase-1 activities in HDL of exercising older individuals. Exp Gerontol 2021; 156:111584. [PMID: 34653558 DOI: 10.1016/j.exger.2021.111584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 09/22/2021] [Accepted: 10/02/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Oxidative stress is an important factor in the formation of atherosclerotic plaques. High-density lipoprotein (HDL) harbors paraoxonase-1 (PON-1) and glutathione peroxidase (GPx), key enzymes in the protection against the harmful effects of oxidative stress. Although exercise training can increase both HDL-c content and its antioxidant action, and glutamine (Gln) intake also promotes GPx-based defenses, the association between exercise training and Gln in the regulation of PON-1 activity was not explored. Therefore, the objective of this study was to investigate the effects of Gln supplementation on the redox balance and on the total HDL antioxidant capacity by evaluation of the activity of PON-1 and GPx enzymes in physically exercised elderly individuals compared to non-exercised ones. METHODS Fifty-one practitioners of a combined exercise training program (CET, age: 71.9 ± 5.7 years) and 32 non-practitioners (NP, age: 73 ± 6.3 years) participated in the study. CET and NP groups were separated into 2 subgroups according to the supplementation: Gln, 0.3 g/kg/day + 10 g maltodextrin (CET-Gln, n = 26; and NP-Gln, n = 16) or placebo, 10 g maltodextrin (CET-PL, n = 25; and NP-PL, n = 16). Blood samples were drawn at baseline and after 30 days after commencement of the supplementation for biochemical and enzyme activity analyses. RESULTS Increased HDL-c, total peroxidase (PRx), and GPx activities were found in both CET-Gln and NP-Gln after the supplementation period, compared to baseline, in opposition to CET-PL and NP-PL groups. PON-1 activity increased only in CET-Gln. In both CET-Gln and NP-Gln groups, there was a reduction of the total peroxides/PRx, iron/PRx, and total peroxides/GPX ratios after supplementation. In CET-Gln, thiobarbituric acid-reactive substances (TBARS)/PRx and TBARS/GPx ratios were also lower after supplementation. CET-Gln and CET-PL subgroups had lower glycemia than NP-Gln and NP-PL, either at baseline or after the supplementation periods. The other parameters were unchanged after supplementation [total cholesterol, LDL-c, triglycerides, non-HDL cholesterol, total peroxides, TBARS, iron serum, Trolox-equivalent antioxidant capacity (TEAC), and uric acid]. CONCLUSIONS Gln supplementation can increase glutathione peroxidase activity regardless the individuals were physically active or sedentary, but the PON-1 activity only increased in physically active individuals. These results show the potential of Gln supplementation in the maintenance of the vascular redox balance, with potential implications for atherogenesis protection.
Collapse
Affiliation(s)
- Renier S Pires
- Post-Graduation Program in Health Sciences, Santo Amaro University (UNISA), São Paulo, SP 04829-300, Brazil
| | - Pedro G S Braga
- Lipid Metabolism Laboratory, Heart Institute (InCor), Medical School Hospital, University of São Paulo, SP 05403-900, Brazil
| | - Juliana M B Santos
- Post-Graduation Program in Science of Human and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP 11015-020, Brazil
| | - Jônatas B Amaral
- ENT Lab, Department of Otorhinolaryngology, Federal University of São Paulo (UNIFESP), São Paulo, SP 04025-002, Brazil
| | - Gislene R Amirato
- ENT Lab, Department of Otorhinolaryngology, Federal University of São Paulo (UNIFESP), São Paulo, SP 04025-002, Brazil
| | - Caio S Trettel
- Interdisciplinary Program in Health Sciences, Institute of Physical Activity Sciences and Sports (ICAFE), Cruzeiro do Sul University, São Paulo, SP 01506-000, Brazil
| | - Carlos A F Dos Santos
- Department of Medicine (Geriatrics and Gerontology), Federal University of São Paulo (UNIFESP), São Paulo, SP 04020-050, Brazil
| | - Mauro Vaisberg
- ENT Lab, Department of Otorhinolaryngology, Federal University of São Paulo (UNIFESP), São Paulo, SP 04025-002, Brazil
| | - Luis H S Nali
- Post-Graduation Program in Health Sciences, Santo Amaro University (UNISA), São Paulo, SP 04829-300, Brazil
| | - Rodolfo P Vieira
- Post-Graduation Program in Science of Human and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, SP 11015-020, Brazil; Post-graduation Program in Bioengineering and Biomedical Engineering, Universidade Brasil, São Paulo, SP 08230-030, Brazil; Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), São Jose dos Campos, SP 12245-520, Brazil
| | - Raul C Maranhão
- Lipid Metabolism Laboratory, Heart Institute (InCor), Medical School Hospital, University of São Paulo, SP 05403-900, Brazil
| | - Tania C Pithon-Curi
- Interdisciplinary Program in Health Sciences, Institute of Physical Activity Sciences and Sports (ICAFE), Cruzeiro do Sul University, São Paulo, SP 01506-000, Brazil
| | - Marcelo P Barros
- Interdisciplinary Program in Health Sciences, Institute of Physical Activity Sciences and Sports (ICAFE), Cruzeiro do Sul University, São Paulo, SP 01506-000, Brazil
| | - André L L Bachi
- Post-Graduation Program in Health Sciences, Santo Amaro University (UNISA), São Paulo, SP 04829-300, Brazil; ENT Lab, Department of Otorhinolaryngology, Federal University of São Paulo (UNIFESP), São Paulo, SP 04025-002, Brazil; Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), São Jose dos Campos, SP 12245-520, Brazil.
| |
Collapse
|
19
|
Pérez-Ruiz I, Ruiz-Sanz JI, Hérnandez ML, Navarro R, Ferrando M, Larreategui Z, Ruiz-Larrea MB. Evidence of Paraoxonases 1, 2, and 3 Expression in Human Ovarian Granulosa Cells. Antioxidants (Basel) 2021; 10:antiox10101504. [PMID: 34679639 PMCID: PMC8532983 DOI: 10.3390/antiox10101504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/05/2021] [Accepted: 09/20/2021] [Indexed: 12/23/2022] Open
Abstract
Increasing evidence suggests that the antioxidant paraoxonase proteins, PON1, PON2, and PON3, have a role in reproduction and may be synthesized by ovarian cells. The aim of this work was to investigate whether human ovarian granulosa cells (GC) express paraoxonases 1, 2, and 3 (PON1, PON2, and PON3) at both the transcriptional and protein levels. Cells were purified from follicle samples of women undergoing ovarian stimulation at oocyte retrieval. We analyzed mRNA by polymerase chain reaction using specific primers for the different variants and quantified the proteins by Western blot using commercially available human recombinant PON proteins as standards. The protein subcellular distribution was determined by immunofluorescence and confocal microscopy and the cell cycles by flow cytometry. Thymidine was used for cellular synchronization at G1/S. Human hepatoma HepG2 and immortalized granulosa COV434 cell lines were used to optimize methodologies. mRNAs from PON1, the two variants of PON2, and PON3 were detected in GC. The cells actively secreted PON1 and PON3, as evidenced by the protein detection in the incubation medium. PON1 and PON3 were mainly distributed in the cytoplasm and notably in the nucleus, while PON2 colocalized with mitochondria. Subcellular nucleo-cytoplasmic distribution of PON1 was associated with the cell cycle. This is the first evidence describing the presence of mRNAs and proteins of the three members of the PON family in human ovarian GC. This study provides the basis of further research to understand the role of these proteins in GC, which will contribute to a better understanding of the reproduction process.
Collapse
Affiliation(s)
- Irantzu Pérez-Ruiz
- Free Radicals and Oxidative Stress (FROS) Research Group of the Department of Physiology, Medicine and Nursing School, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (I.P.-R.); (J.-I.R.-S.); (M.-L.H.); (R.N.)
- BioCruces Health Research Institute, Plaza de Cruces s/n, 48903 Barakaldo, Spain
| | - José-Ignacio Ruiz-Sanz
- Free Radicals and Oxidative Stress (FROS) Research Group of the Department of Physiology, Medicine and Nursing School, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (I.P.-R.); (J.-I.R.-S.); (M.-L.H.); (R.N.)
- BioCruces Health Research Institute, Plaza de Cruces s/n, 48903 Barakaldo, Spain
| | - María-Luisa Hérnandez
- Free Radicals and Oxidative Stress (FROS) Research Group of the Department of Physiology, Medicine and Nursing School, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (I.P.-R.); (J.-I.R.-S.); (M.-L.H.); (R.N.)
- BioCruces Health Research Institute, Plaza de Cruces s/n, 48903 Barakaldo, Spain
| | - Rosaura Navarro
- Free Radicals and Oxidative Stress (FROS) Research Group of the Department of Physiology, Medicine and Nursing School, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (I.P.-R.); (J.-I.R.-S.); (M.-L.H.); (R.N.)
- BioCruces Health Research Institute, Plaza de Cruces s/n, 48903 Barakaldo, Spain
| | - Marcos Ferrando
- Valencian Institute of Infertility (IVI-RMA)-Bilbao, 48940 Leioa, Spain; (M.F.); (Z.L.)
| | - Zaloa Larreategui
- Valencian Institute of Infertility (IVI-RMA)-Bilbao, 48940 Leioa, Spain; (M.F.); (Z.L.)
| | - María-Begoña Ruiz-Larrea
- Free Radicals and Oxidative Stress (FROS) Research Group of the Department of Physiology, Medicine and Nursing School, University of the Basque Country UPV/EHU, 48940 Leioa, Spain; (I.P.-R.); (J.-I.R.-S.); (M.-L.H.); (R.N.)
- BioCruces Health Research Institute, Plaza de Cruces s/n, 48903 Barakaldo, Spain
- Correspondence: ; Tel.: +34-946-012-829
| |
Collapse
|
20
|
Kianmehr A, Qujeq D, Bagheri A, Mahrooz A. Oxidized LDL-regulated microRNAs for evaluating vascular endothelial function: molecular mechanisms and potential biomarker roles in atherosclerosis. Crit Rev Clin Lab Sci 2021; 59:40-53. [PMID: 34523391 DOI: 10.1080/10408363.2021.1974334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
As a simple monolayer, vascular endothelial cells can respond to physicochemical stimuli. In addition to promoting the formation of foam cells, oxidized low-density lipoprotein (ox-LDL) contributes to the atherosclerotic process through different mechanisms, including endothelial cell dysfunction. As conserved noncoding RNAs, microRNAs (miRNAs) naturally lie in different genomic positions and post-transcriptionally regulate the expression of many genes. They participate in integrated networks formed under stress to maintain cellular homeostasis, vascular inflammation, and metabolism. These small RNAs constitute therapeutic targets in different diseases, including atherosclerosis, and their role as biomarkers is crucial given their detectability even years before the emergence of diseases. This review was performed to investigate the role of ox-LDL-regulated miRNAs in atherosclerosis, their molecular mechanisms, and their application as biomarkers of vascular endothelial cell dysfunction.
Collapse
Affiliation(s)
- Anvarsadat Kianmehr
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abouzar Bagheri
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdolkarim Mahrooz
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
21
|
Mishan MA, Khazeei Tabari MA, Mahrooz A, Bagheri A. Role of microRNAs in the anticancer effects of the flavonoid luteolin: a systematic review. Eur J Cancer Prev 2021; 30:413-421. [PMID: 33720053 DOI: 10.1097/cej.0000000000000645] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Flavonoids, a broad class of polyphenolic compounds, can potentially have several therapeutic properties in human diseases, including protective effects against oxidative stress, inflammation, cardiovascular disease, diabetes, neurodegenerative disorders, and cancers. Luteolin as a member of flavonoids has been found to exhibit several anticancer properties mainly through cell apoptosis induction, inhibition of invasion, cell proliferation, network formation, and migration. Recent studies have revealed that phytochemicals such as luteolin may exert therapeutic properties through microRNAs (miRNAs or miRs), which have been emerged as important molecules in cancer biology in recent years. miRNAs, as a class of noncoding RNAs, have several important roles in cancer progression or regression. In this review, we aimed to summarize and discuss the role of miRNAs in the luteolin effects on different cancers. This review can be in line with the studies, which have shown that miRNAs may be potential therapeutic targets in cancer treatment.
Collapse
Affiliation(s)
- Mohammad Amir Mishan
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran
| | | | - Abdolkarim Mahrooz
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center
| | - Abouzar Bagheri
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center
- Department of Clinical Biochemistry and Medical Genetics, Gastrointestinal Cancer Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
22
|
Salari N, Kazeminia M, Mansouri K, Hosseinian-Far A, Mohammadi M. The Activity and Polymorphism of the PON1 in Patients with Chronic Liver Disease: a Systematic Review and Meta-analysis. J Gastrointest Cancer 2021; 53:745-755. [PMID: 34467517 DOI: 10.1007/s12029-021-00699-7] [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] [Accepted: 08/20/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Liver diseases are among the ten deadliest diseases in the world. Measuring PON1 is a test to assess the degree of liver disorder. There are several preliminary studies on the rate of PON1 activity in people with liver disease, and there are differences between the results of these studies; therefore, the aim of this research work is to determine the level of PON1 activity in people with liver disease using meta-analysis. METHOD The study searched to select articles that were published electronically from 2002 to 2020, in national and international databases of SID, MagIran, Embase, ScienceDirect, Scopus, PubMed, and Web of Science (WoS). RESULTS Among the articles included in the meta-analysis, the samples in the case (patients) and control groups were 807 and 2276, respectively. The mean activity of PON1 in individuals with liver disease in the case and control groups were 142.06 ± 7.7 and 272.19 ± 39.6, respectively, and this was statistically significant (P < 0.05). The mean difference analysis highlights a difference of - 2.75 ± 0.48 between the patient and control groups, indicating that liver disease significantly reduces PON1 activity. CONCLUSION The results of this study demonstrate that the polynomorphism of the PON1 is associated with an increased risk of liver disease, with lower levels of PON1 activity in people with liver disease than in healthy patients and this decrease was more in patients with liver cirrhosis than in other liver diseases. Given the importance of this gene's activity, studies such as this could provide a promising path for better drug design and treatment in future.
Collapse
Affiliation(s)
- Nader Salari
- Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Kazeminia
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amin Hosseinian-Far
- Department of Business Systems & Operations, University of Northampton, Northampton, UK
| | - Masoud Mohammadi
- Department of Nursing, School of Nursing and Midwifery, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
23
|
A targeted multi-omics approach reveals paraoxonase-1 as a determinant of obesity-associated fatty liver disease. Clin Epigenetics 2021; 13:158. [PMID: 34389043 PMCID: PMC8360816 DOI: 10.1186/s13148-021-01142-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
Background The multifactorial nature of non-alcoholic fatty liver disease cannot be explained solely by genetic factors. Recent evidence revealed that DNA methylation changes take place at proximal promoters within susceptibility genes. This emphasizes the need for integrating multiple data types to provide a better understanding of the disease’s pathogenesis. One such candidate gene is paraoxonase-1 (PON1). Substantial interindividual differences in PON1 are apparent and could influence disease risk later in life. The aim of this study was therefore to determine the different regulatory aspects of PON1 variability and to examine them in relation to the predisposition to obesity-associated fatty liver disease.
Results A targeted multi-omics approach was applied to investigate the interplay between PON1 genetic variants, promoter methylation, expression profile and enzymatic activity in an adult patient cohort with extensive metabolic and hepatic characterisation including liver biopsy. Alterations in PON1 status were shown to correlate with waist-to-hip ratio and relevant features of liver pathology. Particularly, the regulatory polymorphism rs705379:C > T was strongly associated with more severe liver disease. Multivariable data analysis furthermore indicated a significant association of combined genetic and epigenetic PON1 regulation. This identified relationship postulates a role for DNA methylation as a mediator between PON1 genetics and expression, which is believed to further influence liver disease progression via modifications in PON1 catalytic efficiency. Conclusions Our findings demonstrate that vertical data-integration of genetic and epigenetic regulatory mechanisms generated a more in-depth understanding of the molecular basis underlying the development of obesity-associated fatty liver disease. We gained novel insights into how NAFLD classification and outcome are orchestrated, which could not have been obtained by exclusively considering genetic variation. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01142-1.
Collapse
|
24
|
KORUCU B, HACI H, BALİ EB, DERİCİ MK, GUZ G. A possible contributor to erythropoiesis-stimulating agents requirement in hemodialysis: Paraoxonase 1 activity. TURKISH JOURNAL OF INTERNAL MEDICINE 2021. [DOI: 10.46310/tjim.882813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
25
|
Valencia C SY, Isaza M CA, Henao B J, Beltrán A L, Loango N, Landázuri P. Arylesterase activity of paraoxonase 1 (PON1) on HDL 3 and HDL 2: Relationship with Q192R, C-108T, and L55M polymorphisms. Biochem Biophys Rep 2021; 26:100971. [PMID: 33778169 PMCID: PMC7985468 DOI: 10.1016/j.bbrep.2021.100971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/06/2021] [Accepted: 02/22/2021] [Indexed: 12/15/2022] Open
Abstract
Background Controversy exists regarding the role of the subfractions of high-density lipoproteins (HDL2 and HDL3) in cardiovascular disease. The functionality of these particles, and their protective role, is due in part to the paraoxonase 1 (PON1) presence in them. The polymorphisms rs662 (Q192R, A/G), rs854560 (L55 M, T/A), and rs705379 (C-108T) of the PON1 gene have been related to enzyme activity and, with the anti-oxidative capacity of the HDL. The objective was to determine the arylesterase PON1 activity in HDL3 and HDL2 and its relationship with the polymorphisms mentioned, in a young population. Methods The polymorphisms were determined through mini-sequencing (SnaPshot). The HDL subpopulations were separated via ionic precipitation, cholesterol was measured with enzymatic methods, and PON1 activity was measured through spectrophotometry. Results The results show that the PON1 polymorphisms do not influence the cholesterol in the HDL. A variation between 40.02 and 43.9 mg/dL was in all the polymorphisms without significant differences. Additionally, PON1 activity in the HDL3 subfractions was greater (62.83 ± 20 kU/L) than with HDL2 (35.8 ± 20.8 kU/L) in the whole population and in all the polymorphisms (p < 0.001), and it was independent of the polymorphism and differential arylesterase activity in the Q192R polymorphism (QQ > QR > RR). Thus, 115.90 ± 30.7, 88.78 ± 21.3, 65.29 ± 10.2, respectively, for total HDL, with identical behavior for HDL3 and HDL2. Conclusions PON1 polymorphisms do not influence the HDL-c, and the PON activity is greater in the HDL3 than in the HDL2, independent of the polymorphism, but it is necessary to delve into the functionality of these findings in different populations. PON1 polymorphisms do not influence the cholesterol in the HDL subfractions. PON1 arylesterase activity in the HDL3 subfractions was greater than with HDL2. In Q192R, L55 M and C-108T polymorphisms, PON1 activity is always higher in HDL3. This study shows that healthy young people in Colombia have very low HDL cholesterol.
Collapse
Affiliation(s)
- Sandra Y Valencia C
- Faculty of Health Sciences, Program of Nutrition, Universidad Libre, Colombia.,Faculty of Medicine, Fundación Universitaria Autónoma de las Américas, Colombia.,Faculty of Health Sciences, Universidad del Quindío, Colombia
| | - Carlos A Isaza M
- Faculty of Health Sciences, Universidad Tecnológica de Pereira, Colombia
| | - Julieta Henao B
- Faculty of Health Sciences, Universidad Tecnológica de Pereira, Colombia
| | - Leonardo Beltrán A
- Faculty of Health Sciences, Universidad Tecnológica de Pereira, Colombia.,Faculty of Health Sciences, Unidad Central del Valle del Cauca, Colombia
| | - Nelsy Loango
- Faculty of Health Sciences, Universidad del Quindío, Colombia.,Faculty of Basic Sciences and Technologies, Universidad del Quindío, Colombia
| | | |
Collapse
|
26
|
Kianmehr A, Faraoni I, Kucuk O, Mahrooz A. Epigenetic alterations and genetic variations of angiotensin-converting enzyme 2 (ACE2) as a functional receptor for SARS-CoV-2: potential clinical implications. Eur J Clin Microbiol Infect Dis 2021; 40:1587-1598. [PMID: 33939044 PMCID: PMC8091148 DOI: 10.1007/s10096-021-04264-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 04/27/2021] [Indexed: 02/06/2023]
Abstract
Receptor recognition is a crucial step in viral infection and is a critical factor for cell entry and tissue tropism. In several recent studies, angiotensin-converting enzyme 2 (ACE2) has been demonstrated to be the cellular receptor of SARS-CoV-2 as it was previously well known as the receptor of SARS-CoV. SARS-CoV-2 can bind with high affinity to human ACE2 and engages it as an entry receptor. It seems that the genetic, notably epigenetic variations of ACE2 are less known in different populations, indicating the need for its further investigation. These variations have the potential to affect its contribution to the pathogenicity of COVID-19. The contribution of epigenetics in the interindividual variability of ACE2 merits more attention because epigenetic processes can play important roles in ACE2 alterations in various tissues and different people and populations. Analyzing different DNA methylation patterns and microRNAs, contributing to the ACE2 modulation in the lungs will have a high priority. The epigenetic and genetic variations of ACE2 become even more important when considering that some people have mild clinical symptoms despite having COVID-19. The pathogenicity of SARS-CoV-2 infection is complex; therefore, a better understanding of the underlying pathobiology, especially binding the virus to its receptors, could help improve therapeutic and preventive approaches. This review aims to highlight the importance of evaluating both the epigenetic and genetic variations of ACE2 as a receptor for the deadly SARS-CoV-2.
Collapse
Affiliation(s)
- Anvarsadat Kianmehr
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran ,Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Isabella Faraoni
- Department of Systems Medicine, University of Rome Tor Vergata, 00173 Rom, Italy
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA USA
| | - Abdolkarim Mahrooz
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran ,Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Km 17 Khazarabad Road, Sari, Iran
| |
Collapse
|
27
|
Starling AP, Liu C, Shen G, Yang IV, Kechris K, Borengasser SJ, Boyle KE, Zhang W, Smith HA, Calafat AM, Hamman RF, Adgate JL, Dabelea D. Prenatal Exposure to Per- and Polyfluoroalkyl Substances, Umbilical Cord Blood DNA Methylation, and Cardio-Metabolic Indicators in Newborns: The Healthy Start Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:127014. [PMID: 33356526 PMCID: PMC7759236 DOI: 10.1289/ehp6888] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 05/02/2023]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) are environmentally persistent chemicals widely detected in women of reproductive age. Prenatal PFAS exposure is associated with adverse health outcomes in children. We hypothesized that DNA methylation changes may result from prenatal PFAS exposure and may be linked to offspring cardio-metabolic phenotype. OBJECTIVES We estimated associations of prenatal PFAS with DNA methylation in umbilical cord blood. We evaluated associations of methylation at selected sites with neonatal cardio-metabolic indicators. METHODS Among 583 mother-infant pairs in a prospective cohort, five PFAS were quantified in maternal serum (median 27 wk of gestation). Umbilical cord blood DNA methylation was evaluated using the Illumina HumanMethylation450 array. Differentially methylated positions (DMPs) were evaluated at a false discovery rate ( FDR ) < 0.05 and differentially methylated regions (DMRs) were identified using comb-p (Šidák-adjusted p < 0.05 ). We estimated associations between methylation at candidate DMPs and DMR sites and the following outcomes: newborn weight, adiposity, and cord blood glucose, insulin, lipids, and leptin. RESULTS Maternal serum PFAS concentrations were below the median for females in the U.S. general population. Moderate to high pairwise correlations were observed between PFAS concentrations (ρ = 0.28 - 0.76 ). Methylation at one DMP (cg18587484), annotated to the gene TJAP1, was associated with perfluorooctanoate (PFOA) at FDR < 0.05 . Comb-p detected between 4 and 15 DMRs for each PFAS. Associated genes, some common across multiple PFAS, were implicated in growth (RPTOR), lipid homeostasis (PON1, PON3, CIDEB, NR1H2), inflammation and immune activity (RASL11B, RNF39), among other functions. There was suggestive evidence that two PFAS-associated loci (cg09093485, cg09637273) were associated with cord blood triglycerides and birth weight, respectively (FDR < 0.1 ). DISCUSSION DNA methylation in umbilical cord blood was associated with maternal serum PFAS concentrations during pregnancy, suggesting potential associations with offspring growth, metabolism, and immune function. Future research should explore whether DNA methylation changes mediate associations between prenatal PFAS exposures and child health outcomes. https://doi.org/10.1289/EHP6888.
Collapse
Affiliation(s)
- Anne P. Starling
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cuining Liu
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Guannan Shen
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Ivana V. Yang
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, USA
| | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Sarah J. Borengasser
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kristen E. Boyle
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Weiming Zhang
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Harry A. Smith
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Antonia M. Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Richard F. Hamman
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - John L. Adgate
- Department of Environmental and Occupational Health, Colorado School of Public Health, Aurora, Colorado, USA
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
28
|
Current Understanding of the Relationship of HDL Composition, Structure and Function to Their Cardioprotective Properties in Chronic Kidney Disease. Biomolecules 2020; 10:biom10091348. [PMID: 32967334 PMCID: PMC7564231 DOI: 10.3390/biom10091348] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/27/2022] Open
Abstract
In the general population, the ability of high-density lipoproteins (HDLs) to promote cholesterol efflux is a predictor of cardiovascular events, independently of HDL cholesterol levels. Although patients with chronic kidney disease (CKD) have a high burden of cardiovascular morbidity and mortality, neither serum levels of HDL cholesterol, nor cholesterol efflux capacity associate with cardiovascular events. Important for the following discussion on the role of HDL in CKD is the notion that traditional atherosclerotic cardiovascular risk factors only partially account for this increased incidence of cardiovascular disease in CKD. As a potential explanation, across the spectrum of cardiovascular disease, the relative contribution of atherosclerotic cardiovascular disease becomes less important with advanced CKD. Impaired renal function directly affects the metabolism, composition and functionality of HDL particles. HDLs themselves are a heterogeneous population of particles with distinct sizes and protein composition, all of them affecting the functionality of HDL. Therefore, a more specific approach investigating the functional and compositional features of HDL subclasses might be a valuable strategy to decipher the potential link between HDL, cardiovascular disease and CKD. This review summarizes the current understanding of the relationship of HDL composition, metabolism and function to their cardio-protective properties in CKD, with a focus on CKD-induced changes in the HDL proteome and reverse cholesterol transport capacity. We also will highlight the gaps in the current knowledge regarding important aspects of HDL biology.
Collapse
|
29
|
Mishan MA, Khazeei Tabari MA, Zargari M, Bagheri A. MicroRNAs in the anticancer effects of celecoxib: A systematic review. Eur J Pharmacol 2020; 882:173325. [PMID: 32615181 DOI: 10.1016/j.ejphar.2020.173325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023]
Abstract
Cyclooxygenase-2 (COX-2) is known as an important enzyme in the inflammation process that has tumorigenesis function in various cancers through the induction of epithelial-to-mesenchymal transition (EMT), cell proliferation, migration, and invasion that lead to metastasis. Celecoxib is a nonsteroidal anti-inflammatory drug (NSAID) that can selectively target COX-2, suppress downstream pathways, and finally lead to anticancer potentiality. microRNAs (miRNAs), as a class of small noncoding RNAs, play pivotal roles in cancers through the tumor-suppressive or oncogenic effects, by post-transcriptional regulation of their target genes. In this regard, shreds of evidence have shown that, COX-2 reveals its action through miRNA regulation. So, in this systematic review, we aimed to highlight the tumorigenic role of COX-2 in cancer development and the therapeutic effects of celecoxib, as a selective COX-2 drug, through the regulation of miRNAs.
Collapse
Affiliation(s)
- Mohammad Amir Mishan
- Ocular Tissue Engineering Research Center, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran; USERN Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mehryar Zargari
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abouzar Bagheri
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Studies have shown the three-member paraoxonase (PON) multigene family to be involved in the development of a large variety of diseases with an inflammatory component. Environmental factors such as lifestyle-related factors differ widely between populations and it is important to consider that their impacts may be exerted through the epigenetic mechanisms, which connect genes, the environment and disease development and are a potential therapeutic avenue. RECENT FINDINGS In the review period, very little was published on epigenetics of PON2 or PON3, mostly on their diagnostic value in cancer by measuring methylation levels of these genes. However, the picture is more promising with PON1. Here, several studies have linked the epigenetic regulation of PON1 to various metabolic processes and particularly to the development of several diseases, including stroke, heart disease, aortic valve stenosis and chronic obstructive pulmonary disease. SUMMARY Studies into the epigenetic regulation of the PON family are in their infancy. However, recent studies linking epigenetic regulation of PON1 to disease development will encourage further research and open up the possibility for new potential therapeutic interventions.
Collapse
Affiliation(s)
- Abdolkarim Mahrooz
- Molecular and Cell Biology Research Centre, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | |
Collapse
|
31
|
Mishan MA, Tabari MAK, Parnian J, Fallahi J, Mahrooz A, Bagheri A. Functional mechanisms of miR-192 family in cancer. Genes Chromosomes Cancer 2020; 59:722-735. [PMID: 32706406 DOI: 10.1002/gcc.22889] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/14/2020] [Accepted: 07/21/2020] [Indexed: 12/11/2022] Open
Abstract
By growing research on the mechanisms and functions of microRNAs (miRNAs, miRs), the role of these noncoding RNAs gained more attention in healthcare. Due to the remarkable regulatory role of miRNAs, any dysregulation in their expression causes cellular functional impairment. In recent years, it has become increasingly apparent that these small molecules contribute to development, cell differentiation, proliferation, apoptosis, and tumor growth. In many studies, the miR-192 family has been suggested as a potential prognostic and diagnostic biomarker and even as a possible therapeutic target for several cancers. However, the mechanistic effects of the miR-192 family on cancer cells are still controversial. Here, we have reviewed each family member of the miR-192 including miR-192, miR-194, and miR-215, and discussed their mechanistic roles in various cancers.
Collapse
Affiliation(s)
- Mohammad Amir Mishan
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
- USERN Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Javad Parnian
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Jafar Fallahi
- Molecular Medicine Department, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abdolkarim Mahrooz
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abouzar Bagheri
- Department of Clinical Biochemistry and Medical Genetics, Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
32
|
Shokri Y, Variji A, Nosrati M, Khonakdar-Tarsi A, Kianmehr A, Kashi Z, Bahar A, Bagheri A, Mahrooz A. Importance of paraoxonase 1 (PON1) as an antioxidant and antiatherogenic enzyme in the cardiovascular complications of type 2 diabetes: Genotypic and phenotypic evaluation. Diabetes Res Clin Pract 2020; 161:108067. [PMID: 32044348 DOI: 10.1016/j.diabres.2020.108067] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/13/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022]
Abstract
Oxidant-antioxidant imbalance is involved in the etiology of different diseases, including cardiovascular diseases (CVDs), liver disorders, kidney diseases, cancers and diabetes mellitus. Antioxidant enzymes play a key role in striking an oxidant-antioxidant balance. Moreover, paraoxonase 1 (PON1) is an antioxidant enzyme that binds with high-density lipoprotein (HDL) in the circulation, and antioxidant and antiaterogenic properties of this lipoprotein are significantly associated with PON1. Research suggests PON1 contributes to the pathogenesis of certain human diseases such as type 2 diabetes (T2D). The association between PON1 and T2D appear to be reciprocal so that the disease significantly decreases PON1 levels and in turn, the genetics of PON1 may have a role the risk of susceptibility to T2D. Several factors that reduce the activity and concentration of PON1 in patients with T2D include increased glycation and loss-of-function polymorphisms. The genotypic and phenotypic evaluations of PON1 are therefore crucial for assessing the risk of cardiovascular complications in these patients, and strategies for increasing or restoring PON1 levels are useful for reducing or preventing their cardiovascular complications as their main cause of mortality. The present review aimed at discussing and emphasizing the key role of PON1 in T2D as a silent and dangerous disease.
Collapse
Affiliation(s)
- Yasaman Shokri
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Atena Variji
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mani Nosrati
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abbas Khonakdar-Tarsi
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Anvarsadat Kianmehr
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan, Iran; Department of Medical Biotechnology, Faculty of Advanced Madical Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zahra Kashi
- Diabetes Research Center, Imam Teaching Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Adele Bahar
- Diabetes Research Center, Imam Teaching Hospital, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abouzar Bagheri
- Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Abdolkarim Mahrooz
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Diabetes Research Center, Imam Teaching Hospital, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
33
|
Farràs M, Canyelles M, Fitó M, Escolà-Gil JC. Effects of Virgin Olive Oil and Phenol-Enriched Virgin Olive Oils on Lipoprotein Atherogenicity. Nutrients 2020; 12:nu12030601. [PMID: 32110861 PMCID: PMC7146215 DOI: 10.3390/nu12030601] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 01/22/2023] Open
Abstract
The atherogenicity of low-density lipoprotein (LDL) and triglyceride-rich lipoproteins (TRLs) may be more significant than LDL cholesterol levels. Clinical trials which have led to increased high-density lipoprotein (HDL) cholesterol have not always seen reductions in cardiovascular disease (CVD). Furthermore, genetic variants predisposing individuals to high HDL cholesterol are not associated with a lower risk of suffering a coronary event, and therefore HDL functionality is considered to be the most relevant aspect. Virgin olive oil (VOO) is thought to play a protective role against CVD. This review describes the effects of VOO and phenol-enriched VOOs on lipoprotein atherogenicity and HDL atheroprotective properties. The studies have demonstrated a decrease in LDL atherogenicity and an increase in the HDL-mediated macrophage cholesterol efflux capacity, HDL antioxidant activity, and HDL anti-inflammatory characteristics after various VOO interventions. Moreover, the expression of cholesterol efflux-related genes was enhanced after exposure to phenol-enriched VOOs in both post-prandial and sustained trials. Improvements in HDL antioxidant properties were also observed after VOO and phenol-enriched VOO interventions. Furthermore, some studies have demonstrated improved characteristics of TRL atherogenicity under postprandial conditions after VOO intake. Large-scale, long-term randomized clinical trials, and Mendelian analyses which assess the lipoprotein state and properties, are required to confirm these results.
Collapse
Affiliation(s)
- Marta Farràs
- Molecular Bases of Cardiovascular Risk Group Institut de Recerca de l’Hospital Santa Creu i Sant Pau-Institut d’Investigacions Biomèdiques (IIB) Sant Pau, 08041 Barcelona, Spain; (M.C.); (J.C.E.-G.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-935537595
| | - Marina Canyelles
- Molecular Bases of Cardiovascular Risk Group Institut de Recerca de l’Hospital Santa Creu i Sant Pau-Institut d’Investigacions Biomèdiques (IIB) Sant Pau, 08041 Barcelona, Spain; (M.C.); (J.C.E.-G.)
- Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Departament de Bioquímica, Biologia Molecular i Biomedicina, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Montserrat Fitó
- Cardiovascular Risk and Nutrition Research Group, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain;
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 28029 Madrid, Spain
| | - Joan Carles Escolà-Gil
- Molecular Bases of Cardiovascular Risk Group Institut de Recerca de l’Hospital Santa Creu i Sant Pau-Institut d’Investigacions Biomèdiques (IIB) Sant Pau, 08041 Barcelona, Spain; (M.C.); (J.C.E.-G.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, 28029 Madrid, Spain
- Departament de Bioquímica, Biologia Molecular i Biomedicina, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
34
|
Milaciu MV, Vesa ȘC, Bocșan IC, Ciumărnean L, Sâmpelean D, Negrean V, Pop RM, Matei DM, Pașca S, Răchișan AL, Buzoianu AD, Acalovschi M. Paraoxonase-1 Serum Concentration and PON1 Gene Polymorphisms: Relationship with Non-Alcoholic Fatty Liver Disease. J Clin Med 2019; 8:jcm8122200. [PMID: 31847187 PMCID: PMC6947206 DOI: 10.3390/jcm8122200] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/08/2019] [Accepted: 12/11/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Non-alcoholic fatty liver disease (NAFLD) is an important cause of chronic liver diseases around the world. Paraoxonase-1 (PON1) is an enzyme produced by the liver with an important antioxidant role. The aim of this study was to evaluate PON1 serum concentration and PON1 gene polymorphisms in patients with NAFLD. Materials and methods: We studied a group of 81 patients with NAFLD with persistently elevated aminotransferases and a control group of 81 patients without liver diseases. We collected clinical information and performed routine blood tests. We also measured the serum concentration of PON1 and evaluated the PON1 gene polymorphisms L55M, Q192R, and C-108T. Results: There was a significant difference (p < 0.001) in serum PON1 concentrations among the two groups. The heterozygous and the mutated homozygous variants (LM + MM) of the L55M polymorphism were more frequent in the NAFLD group (p < 0.001). These genotypes were found in a multivariate binary logistic regression to be independently linked to NAFLD (Odds ratio = 3.4; p = 0.04). In a multivariate linear regression model, the presence of NAFLD was associated with low PON1 concentration (p < 0.001). Conclusions: PON1 serum concentrations were diminished in patients with NAFLD, and the presence of NAFLD was linked with low PON1 concentration. The LM + MM genotypes of the PON1 L55M polymorphism were an independent predictor for NAFLD with persistently elevated aminotransferases.
Collapse
Affiliation(s)
- Mircea Vasile Milaciu
- Department 5—Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (M.V.M.); (D.S.); (V.N.)
| | - Ștefan Cristian Vesa
- Department 2—Functional Sciences, Discipline of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (Ș.C.V.); (I.C.B.); (R.M.P.); (A.D.B.)
| | - Ioana Corina Bocșan
- Department 2—Functional Sciences, Discipline of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (Ș.C.V.); (I.C.B.); (R.M.P.); (A.D.B.)
| | - Lorena Ciumărnean
- Department 5—Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (M.V.M.); (D.S.); (V.N.)
- Correspondence:
| | - Dorel Sâmpelean
- Department 5—Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (M.V.M.); (D.S.); (V.N.)
| | - Vasile Negrean
- Department 5—Internal Medicine, 4th Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania; (M.V.M.); (D.S.); (V.N.)
| | - Raluca Maria Pop
- Department 2—Functional Sciences, Discipline of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (Ș.C.V.); (I.C.B.); (R.M.P.); (A.D.B.)
| | - Daniela Maria Matei
- Department 5—Internal Medicine, 3rd Medical Clinic, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania;
| | - Sergiu Pașca
- Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Andreea Liana Răchișan
- Department of Pediatrics, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400177 Cluj-Napoca, Romania;
| | - Anca Dana Buzoianu
- Department 2—Functional Sciences, Discipline of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (Ș.C.V.); (I.C.B.); (R.M.P.); (A.D.B.)
| | - Monica Acalovschi
- Doctoral School, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| |
Collapse
|