1
|
Isah MB, Tajuddeen N, Yusuf A, Mohammed A, Ibrahim MA, Melzig M, Zhang X. The antidiabetic properties of lignans: a comprehensive review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156717. [PMID: 40220408 DOI: 10.1016/j.phymed.2025.156717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Diabetes mellitus (DM) is a chronic metabolic disease with a high global prevalence. Lignans, a class of plant natural compounds found in commonly consumed foods, are well-tolerated by humans and have demonstrated promising potential in the management of DM. Consumption of lignan-rich foods has been associated with improved overall health and quality of life. PURPOSE The clinical and preclinical evidence on the role of lignans in managing DM are critically examined. METHODS A thorough literature search was conducted across major scientific databases, focusing on studies that reported the effects of individual lignans on key diabetes indicators, such as glucose utilisation and insulin sensitivity, in both human and animal models, as well as in cell-based studies. RESULTS A total of 180 lignans were included in the review. Out of these, only three were investigated in randomised clinical trials in humans and 31 in animal models. The reviewed evidence suggests some beneficial effects of lignans in preventing the development of obesity-related diabetes. Their therapeutic benefits in preventing diabetes-related complications, particularly diabetic nephropathy, in both type 1 and type 2 diabetes, are also supported. Metabolites of various lignans, produced by microbial metabolism in the gut and serum enzymes, appear to be key bioactive forms, highlighting the need for detailed pharmacodynamic studies, optimised dosage designs, and the use of the appropriate lignan molecules for cell-based screening. CONCLUSION Lignans and their microbial metabolites show promise in preventing obesity-related diabetes and mitigating diabetes-related complications such as diabetic nephropathy, though further clinical studies are needed to optimize their therapeutic potential.
Collapse
Affiliation(s)
- Murtala Bindawa Isah
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Department of Biochemistry, Umaru Musa Yar'adua University Katsina, Nigeria.
| | - Nasir Tajuddeen
- Department of Chemistry, Ahmadu Bello University Zaria, Nigeria
| | - Anas Yusuf
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China
| | - Aminu Mohammed
- Department of Biochemistry, Ahmadu Bello University Zaria, Nigeria
| | | | - Matthias Melzig
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Freie Universitaet Berlin, Institute of Pharmacy, Berlin, Germany.
| | - Xiaoying Zhang
- Chinese-German Joint Laboratory for Natural Product Research, Shaanxi International Cooperation Demonstration Base, Shaanxi University of Technology, Hanzhong, 723000, Shaanxi, China; Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal; Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
2
|
Yang J, Wu Y, Jiang Z, Jiao L, Wang Y. Fecal fatty acid profile reveals the therapeutic effect of red ginseng acidic polysaccharide on type 2 diabetes mellitus in rats. J Food Sci 2025; 90:e70015. [PMID: 39832227 DOI: 10.1111/1750-3841.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/13/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
This study aimed to investigate the potential hypoglycemic mechanism of red ginseng acidic polysaccharides (RGAP) from the perspective of fatty acid (FA) regulation. A high-glucose/high-fat diet in conjunction with streptozotocin administration was employed to establish type 2 diabetes mellitus (T2DM) rat models, and their fecal FAs were detected using the liquid chromatography-mass spectrometry (LC-MS) method. RGAP treatment alleviated the polyphagia, polydipsia, weight loss, and hyperglycemia observed in T2DM rats. FA profile was disturbed by T2DM modeling, and 11 marker FAs were selected from statistical analysis, whose intensities were reversely changed by RGAP administration. Among these marker FAs, short-chain FAs were negatively correlated with the fasting blood glucose (FBG) level, while positive correlations were observed between long-chain FA and the FBG level. Combined with the metabolite-enzyme-gene network analysis, we inferred that the mechanistic mechanism RGAP on T2DM may be associated with the regulation of FA metabolism and inflammation-related signaling pathways. This study confirmed the regulatory effect of RGAP on fecal FA, which can provide a scientific basis and new ideas for developing red ginseng as a functional food for supplementary treatment of T2DM.
Collapse
Affiliation(s)
- Jingxuan Yang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, China
| | - Yi Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Ziye Jiang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Lili Jiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Yang Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
3
|
Nawaz Z, Riaz N, Saleem M, Iqbal A, Ejaz SA, Muzaffar S, Bashir B, Ashraf M, Rehman AU, Bilal MS, Prabhala BK, Sajid S. Probing N-substituted 4-(5-mercapto-4-ethyl-4H-1,2,4-triazol-3-yl)- N-phenylpiperdine-1-carboxamides as potent 15-LOX inhibitors supported with ADME, DFT calculations and molecular docking studies. Heliyon 2024; 10:e35278. [PMID: 39281606 PMCID: PMC11401107 DOI: 10.1016/j.heliyon.2024.e35278] [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: 03/19/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 09/18/2024] Open
Abstract
In our continuous efforts to find out leads against the enzyme 15-lipoxygenase (15-LOX), the current study deals with the synthesis of a series of new N-alkyl/aralkyl/aryl derivatives of 2-(4-ethyl-5-(1-phenylcarbamoyl)piperidine-4H-1,2,4-triazol-3-ylthio)methylacetamide (7a-n) with anti-LOX activities. The synthesis was started by reacting phenylisocyanate with isonipecotate that sequentially converted into N-substituted ester (1), hydrazide (2), semicarbazide (3) and N-ethylated 5-(1-phenylcarbamoyl)piperidine-1,2,4-triazole (4). The final compounds, 7a-n, were obtained by reacting 4 with various N-alkyl/aralkyl/aryl electrophiles. Both the intermediates and target compounds were characterized by FTIR, 1H, 13C NMR spectroscopy, EI-MS and HR-EI-MS spectrometry and screened against soybean 15-LOX by chemiluminescence method. The eight compounds 7e, 7j, 7h, 7a, 7g, 7b, 7n, 7c showed potent inhibitory activities against 15-LOX with values ranging from IC50 0.36 ± 0.15 μM (7e) to IC50 6.75 ± 0.17 μM (7c) compared with the reference quercetin (IC50 4.86 ± 0.14 μM) and baicalein (IC50 2.24 ± 0.13 μM). Two analogues (7l, 7f) had significantly outstanding inhibitory potential with IC50 values 12.15 ± 0.23 μM and 15.54 ± 0.26 μM, whereas, the derivatives 7i, and 7d displayed IC50 values of 21.56 ± 0.27 μM, 23.59 ± 0.24 μM and the compounds 7k, 7m were found inactive. All analogues exhibited blood mononuclear cells (MNCs) viability >75 % at 0.25 mM concentration as determined by MTT method. Calculated pharmacokinetic properties projected good lipophilicity, bioavailability and drug-likeness properties and did not violate Lipinski's/Veber rule. Molecular docking studies revealed lower binding free energies of all the derivatives than the reference compounds. The binding free energies were -9.8 kcal/mol, -9.70 k/mol and -9.20 kcal/mol for 7j, 7h and 7e, respectively, compared with the standard quercetin (-8.47 kcal/mol) and baicalein (-8.98 kcal/mol). The docked ligands formed hydrogen bonds with the amino acid residues Gln598 (7e), Arg260, Val 126 (7h), Gln762, Gln574, Thr443, Arg580 (7j) while other hydrophobic interactions observed therein further stabilized the complexes. The results of density functional theory (DFT) revealed that analogues with more stabilized lower unoccupied molecular orbital (LUMO) had significant enzyme inhibitory activity. The data collectively supports these molecules as leads against 15-LOX and demand further investigations as anti-inflammatory agents.
Collapse
Affiliation(s)
- Zahid Nawaz
- Institute of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Naheed Riaz
- Institute of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Muhammad Saleem
- Institute of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Ambar Iqbal
- Institute of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
- Department of Biochemistry, Institute of Biochemistry, Biotechnology and Bioinformatics (IBBB), Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Syeda Abida Ejaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Khawaja Fareed Campus, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Saima Muzaffar
- Department of Chemistry, Division of Sceience and Technology, University of Education, 54770, Lahore, Vehari Campus, Pakistan
| | - Bushra Bashir
- Institute of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Muhammad Ashraf
- Institute of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Aziz-Ur Rehman
- Department of Chemistry, Government College University Lahore, Lahore, 54000, Pakistan
| | - Muhammad Sajjad Bilal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Khawaja Fareed Campus, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Bala Krishna Prabhala
- Institute of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230M, Denmark
| | - Salvia Sajid
- Department of Drug Design and Pharmacology, University of Copenhagen 2, DK-2100, Kobenhavn O, Denmark
| |
Collapse
|
4
|
Amin A, Ullah N, Khan MA, Elsadek MF, Elshikh MS, Hasnain SZU, Baloch R, Chaman S, Makhkamov T, Yuldashev A, Yunusov S, Biturku J. Mango peel extracts and mangiferin chromatographic Fourier-transform infrared correlation with antioxidant, antidiabetic, and advanced glycation end product inhibitory potentials using in silico modeling and in vitro assays. Biomed Chromatogr 2024; 38:e5936. [PMID: 38956791 DOI: 10.1002/bmc.5936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/03/2024] [Accepted: 05/31/2024] [Indexed: 07/04/2024]
Abstract
Mangifera indica peels are a rich source of diverse flavonoids and xanthonoids; however, generally these are discarded. Computational studies revealed that mangiferin significantly interacts with amino acid residues of transcriptional regulators 1IK3, 3TOP, and 4f5S. The methanolic extract of Langra variety of mangoes contained the least phenol concentrations (22.6 ± 0.32 mg/gGAE [gallic acid equivalent]) compared to the chloroform (214.8 ± 0.12 mg/gGAE) and ethyl acetate fractions (195.6 ± 0.14 mg/gGAE). Similarly, the methanolic extract of Sindhri variety contained lower phenol concentrations (42.3 ± 0.13 mg/gRUE [relative utilization efficiency]) compared with the chloroform (85.6 ± 0.15 mg/gGAE) and ethyl acetate (76.1 ± 0.32 mg/gGAE) fractions. Langra extract exhibited significant α-glucosidase inhibition (IC50 0.06 mg/mL), whereas the ethyl acetate fraction was highly active (IC50 0.12 mg/mL) in Sindhri variety. Mangiferin exhibited significant inhibition (IC50 0.026 mg/mL). A moderate inhibition of 15-LOX was observed in all samples, whereas mangiferin was least active. In advanced glycation end product inhibition assay, the chloroform fraction of Langra variety exhibited significant inhibition in nonoxidative (IC50 64.4 μg/mL) and oxidative modes (IC50 54.7 μg/mL). It was concluded that both Langra and Sindhri peel extracts and fractions possess significant antidiabetic activities. The results suggest the potential use of peel waste in the management and complications of diabetes.
Collapse
Affiliation(s)
- Adnan Amin
- Natural Products Research Lab, Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, Dera Ismail Khan, Pakistan
| | - Niamat Ullah
- Natural Products Research Lab, Gomal Centre of Pharmaceutical Sciences, Faculty of Pharmacy, Gomal University, Dera Ismail Khan, Pakistan
| | - Mohsin Abbas Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | | | - Mohamed S Elshikh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Syed Zia Ul Hasnain
- Department of Pharmacognosy, Faculty of Pharmacy, Bahauddin Zakaraiya University, Multan, Pakistan
| | - Rabia Baloch
- Allama Iqbal Teaching Hospital, Dera Ghazi Khan, Punjab, Pakistan
| | - Sadia Chaman
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Punjab, Pakistan
| | - Trobjon Makhkamov
- Department of Forestry and Landscape Design, Tashkent State Agrarian University, Tashkent, Uzbekistan
| | - Akramjon Yuldashev
- Department of Ecology and Botany, Andijan State University, Andijan, Uzbekistan
| | - Salohiddinjon Yunusov
- Department of Horticulture and Viticulture, Tashkent State Agrarian University, Tashkent, Uzbekistan
| | - Jonida Biturku
- Department of Agronomy Sciences, Faculty of Agriculture and Environment, Agriculture University of Tirana, Tirana, Albania
| |
Collapse
|
5
|
Zhao L, Yang W, Ji W, Pan Q, Yang J, Cao X. Untargeted metabolomics uncovers metabolic dysregulation and tissue sensitivity in ACE2 knockout mice. Heliyon 2024; 10:e27472. [PMID: 38496880 PMCID: PMC10944221 DOI: 10.1016/j.heliyon.2024.e27472] [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: 12/04/2023] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) polymorphisms are associated with increased risk of type 2 diabetes mellitus (T2DM), obesity and dyslipidemia, which have been determined in various populations. Consistently, ACE2 knockout (ACE2 KO) mice display damaged energy metabolism in multiple tissues, especially the key metabolic tissues such as liver, skeletal muscle and epididymal white adipose tissue (eWAT) and show even more severe phenotype under high-fat diet (HFD) induced metabolic stress. However, the effects of ACE2 on global metabolomics profiling and the tissue sensitivity remain unclear. To understand how tissues independently and collectively respond to ACE2, we performed untargeted metabolomics in serum in ACE2 KO and control wild type (WT) mice both on normal diet (ND) and HFD, and in three key metabolic tissues (liver, skeletal muscle and eWAT) after HFD treatment. The results showed significant alterations in metabolic profiling in ACE2 KO mice. We identified 275 and 168 serum metabolites differing significantly between WT and ACE2 KO mice fed on ND and HFD, respectively. And the altered metabolites in the ACE2 KO group varied from 90 to 196 in liver, muscle and eWAT. The alterations in ND and HFD serum were most similar. Compared with WT mice, ACE2 KO mice showed an increase in N-phenylacetylglutamine (PAGln), methyl indole-3-acetate, 5-hydroxytryptophol, cholic acid, deoxycholic acid and 12(S)-HETE, while LPC (19:0) and LPE (16:1) decreased. Moreover, LPC (20:0), LPC (20:1) and PC (14:0e/6:0) were reduced in both ND and HFD serum, paralleling the decreases identified in HFD skeletal muscle. Interestingly, DL-tryptophan, indole and Gly-Phe decreased in both ND and HFD serum but were elevated in HFD liver of ACE2 KO mice. A low level of l-ergothioneine was observed among liver, muscle, and epididymal fat tissue of ACE2 KO mice. Pathway analysis demonstrated that different tissues exhibited different dysregulated metabolic pathways. In conclusion, these results revealed that ACE2 deficiency leads to an overall state of metabolic distress, which may provide a new insight into the underlying pathogenesis in metabolic disorders in both ACE2 KO mice and in patients with certain genetic variant of ACE2 gene.
Collapse
Affiliation(s)
| | | | - Wenyi Ji
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Qiuyue Pan
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Jinkui Yang
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xi Cao
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| |
Collapse
|
6
|
Li XJ, Suo P, Wang YN, Zou L, Nie XL, Zhao YY, Miao H. Arachidonic acid metabolism as a therapeutic target in AKI-to-CKD transition. Front Pharmacol 2024; 15:1365802. [PMID: 38523633 PMCID: PMC10957658 DOI: 10.3389/fphar.2024.1365802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 03/26/2024] Open
Abstract
Arachidonic acid (AA) is a main component of cell membrane lipids. AA is mainly metabolized by three enzymes: cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (CYP450). Esterified AA is hydrolysed by phospholipase A2 into a free form that is further metabolized by COX, LOX and CYP450 to a wide range of bioactive mediators, including prostaglandins, lipoxins, thromboxanes, leukotrienes, hydroxyeicosatetraenoic acids and epoxyeicosatrienoic acids. Increased mitochondrial oxidative stress is considered to be a central mechanism in the pathophysiology of the kidney. Along with increased oxidative stress, apoptosis, inflammation and tissue fibrosis drive the progressive loss of kidney function, affecting the glomerular filtration barrier and the tubulointerstitium. Recent studies have shown that AA and its active derivative eicosanoids play important roles in the regulation of physiological kidney function and the pathogenesis of kidney disease. These factors are potentially novel biomarkers, especially in the context of their involvement in inflammatory processes and oxidative stress. In this review, we introduce the three main metabolic pathways of AA and discuss the molecular mechanisms by which these pathways affect the progression of acute kidney injury (AKI), diabetic nephropathy (DN) and renal cell carcinoma (RCC). This review may provide new therapeutic targets for the identification of AKI to CKD continuum.
Collapse
Affiliation(s)
- Xiao-Jun Li
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ping Suo
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yan-Ni Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Liang Zou
- School of Food and Bioengineering, Chengdu University, Chengdu, Sichuan, China
| | - Xiao-Li Nie
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying-Yong Zhao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Hua Miao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Sarkar S, Deiter C, Kyle JE, Guney MA, Sarbaugh D, Yin R, Li X, Cui Y, Ramos-Rodriguez M, Nicora CD, Syed F, Juan-Mateu J, Muralidharan C, Pasquali L, Evans-Molina C, Eizirik DL, Webb-Robertson BJM, Burnum-Johnson K, Orr G, Laskin J, Metz TO, Mirmira RG, Sussel L, Ansong C, Nakayasu ES. Regulation of β-cell death by ADP-ribosylhydrolase ARH3 via lipid signaling in insulitis. Cell Commun Signal 2024; 22:141. [PMID: 38383396 PMCID: PMC10880366 DOI: 10.1186/s12964-023-01437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/12/2023] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Lipids are regulators of insulitis and β-cell death in type 1 diabetes development, but the underlying mechanisms are poorly understood. Here, we investigated how the islet lipid composition and downstream signaling regulate β-cell death. METHODS We performed lipidomics using three models of insulitis: human islets and EndoC-βH1 β cells treated with the pro-inflammatory cytokines interlukine-1β and interferon-γ, and islets from pre-diabetic non-obese mice. We also performed mass spectrometry and fluorescence imaging to determine the localization of lipids and enzyme in islets. RNAi, apoptotic assay, and qPCR were performed to determine the role of a specific factor in lipid-mediated cytokine signaling. RESULTS Across all three models, lipidomic analyses showed a consistent increase of lysophosphatidylcholine species and phosphatidylcholines with polyunsaturated fatty acids and a reduction of triacylglycerol species. Imaging assays showed that phosphatidylcholines with polyunsaturated fatty acids and their hydrolyzing enzyme phospholipase PLA2G6 are enriched in islets. In downstream signaling, omega-3 fatty acids reduce cytokine-induced β-cell death by improving the expression of ADP-ribosylhydrolase ARH3. The mechanism involves omega-3 fatty acid-mediated reduction of the histone methylation polycomb complex PRC2 component Suz12, upregulating the expression of Arh3, which in turn decreases cell apoptosis. CONCLUSIONS Our data provide insights into the change of lipidomics landscape in β cells during insulitis and identify a protective mechanism by omega-3 fatty acids. Video Abstract.
Collapse
Affiliation(s)
- Soumyadeep Sarkar
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Cailin Deiter
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Center, Aurora, CO, 80045, USA
| | - Jennifer E Kyle
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Michelle A Guney
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Center, Aurora, CO, 80045, USA
| | - Dylan Sarbaugh
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Center, Aurora, CO, 80045, USA
| | - Ruichuan Yin
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907-2084, USA
| | - Xiangtang Li
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907-2084, USA
| | - Yi Cui
- Environmental and Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
- NanoString Technologies, Seattle, WA, 98109, USA
| | - Mireia Ramos-Rodriguez
- Endocrine Regulatory Genomics, Department of Experimental & Health Sciences, University Pompeu Fabra, 08003, Barcelona, Spain
| | - Carrie D Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Farooq Syed
- Center for Diabetes and Metabolic Diseases and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jonas Juan-Mateu
- ULB Center for Diabetes Research, Université Libre de Bruxelles (ULB), 1070, Brussels, Belgium
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003, Barcelona, Spain
| | - Charanya Muralidharan
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Lorenzo Pasquali
- Endocrine Regulatory Genomics, Department of Experimental & Health Sciences, University Pompeu Fabra, 08003, Barcelona, Spain
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles (ULB), 1070, Brussels, Belgium
| | - Bobbie-Jo M Webb-Robertson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Center, Aurora, CO, 80045, USA
| | - Kristin Burnum-Johnson
- Environmental and Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Galya Orr
- Environmental and Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Julia Laskin
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907-2084, USA
| | - Thomas O Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Raghavendra G Mirmira
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Lori Sussel
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Center, Aurora, CO, 80045, USA
| | - Charles Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
| | - Ernesto S Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99354, USA.
| |
Collapse
|
8
|
Safdar NZ, Kietsiriroje N, Ajjan RA. The Cellular and Protein Arms of Coagulation in Diabetes: Established and Potential Targets for the Reduction of Thrombotic Risk. Int J Mol Sci 2023; 24:15328. [PMID: 37895008 PMCID: PMC10607436 DOI: 10.3390/ijms242015328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Diabetes is a metabolic condition with a rising global prevalence and is characterised by abnormally high blood glucose levels. Cardiovascular disease (CVD) accounts for the majority of deaths in diabetes and, despite improvements in therapy, mortality and hospitalisations in this cohort remain disproportionally higher compared to individuals with normal glucose metabolism. One mechanism for increased CVD risk is enhanced thrombosis potential, due to altered function of the cellular and acellular arms of coagulation. Different mechanisms have been identified that mediate disordered blood clot formation and breakdown in diabetes, including dysglycaemia, insulin resistance, and metabolic co-morbidities. Collectively, these induce platelet/endothelial dysfunction and impair the fibrinolytic process, thus creating a prothrombotic milieu. Despite these abnormalities, current antithrombotic therapies are largely similar in diabetes compared to those without this condition, which explains the high proportion of patients experiencing treatment failure while also displaying an increased risk of bleeding events. In this narrative review, we aimed to summarise the physiological functioning of haemostasis followed by the pathological effects of diabetes mellitus on platelets and the fibrin network. Moreover, we carefully reviewed the literature to describe the current and future therapeutic targets to lower the thrombosis risk and improve vascular outcomes in diabetes.
Collapse
Affiliation(s)
- Nawaz Z. Safdar
- Department of Internal Medicine, St James’s University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds LS9 7TF, UK;
- Light Laboratories, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, 6 Clarendon Way, Leeds LS2 3AA, UK
| | - Noppadol Kietsiriroje
- Endocrinology and Metabolism Unit, Faculty of Medicine, Prince of Songkla University, Songkla 90110, Thailand;
| | - Ramzi A. Ajjan
- Light Laboratories, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, 6 Clarendon Way, Leeds LS2 3AA, UK
| |
Collapse
|
9
|
Rohlik DL, Patel E, Gilbert NC, Offenbacher AR, Garcia BL. Investigating membrane-binding properties of lipoxygenases using surface plasmon resonance. Biochem Biophys Res Commun 2023; 670:47-54. [PMID: 37276790 PMCID: PMC10330842 DOI: 10.1016/j.bbrc.2023.05.066] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/18/2023] [Indexed: 06/07/2023]
Abstract
Lipoxygenases (LOXs) catalyze the oxidation of polyunsaturated fatty acids and synthesize oxylipin products that drive important cellular signaling processes in plants and animals. While there has been indirect evidence presented for the interaction of mammalian LOXs with membranes, a quantitative study of the molecular details of LOX-membrane interactions is lacking. Here, we mimicked biological membranes using surface plasmon resonance (SPR) sensor chips derivatized with 2-D planar lipophilic anchors (2D LP) to capture liposomes of varying phospholipid compositions that self-assemble into lipid bilayers on the SPR chip. The sensor chip surfaces were then used to investigate the membrane-binding properties of model LOX enzymes. SPR binding assays displayed reproducible and stable liposome capture to the sensor chip surface that allowed for the detailed characterization of LOX-membrane interactions. Our studies demonstrate a calcium-dependence for the membrane binding activities of coral 8R-LOX and human 15-LOX-2. Furthermore, our data confirm the importance of key membrane insertion loop residues in each of these LOX enzymes for membrane binding activity. Experiments utilizing model plant and human LOXs reveal differences in membrane-binding specificities. Our study establishes and validates a robust SPR-based platform using 2D LP sensor chips that allows for the detailed study of LOX-membrane interactions under different experimental conditions, including altered membrane compositions. Collectively, this investigation improves our overall understanding of LOX-membrane interaction properties, and our SPR-based approach holds potential for future use in the development of LOX-based therapeutics.
Collapse
Affiliation(s)
- Denise L Rohlik
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Ethan Patel
- Department Chemistry, East Carolina University, Greenville, NC, USA
| | - Nathaniel C Gilbert
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | | | - Brandon L Garcia
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
10
|
Ludovico ID, Sarkar S, Elliott E, Virtanen SM, Erlund I, Ramanadham S, Mirmira RG, Metz TO, Nakayasu ES. Fatty acid-mediated signaling as a target for developing type 1 diabetes therapies. Expert Opin Ther Targets 2023; 27:793-806. [PMID: 37706269 PMCID: PMC10591803 DOI: 10.1080/14728222.2023.2259099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/11/2023] [Indexed: 09/15/2023]
Abstract
INTRODUCTION Type 1 diabetes (T1D) is an autoimmune disease in which pro-inflammatory and cytotoxic signaling drive the death of the insulin-producing β cells. This complex signaling is regulated in part by fatty acids and their bioproducts, making them excellent therapeutic targets. AREAS COVERED We provide an overview of the fatty acid actions on β cells by discussing how they can cause lipotoxicity or regulate inflammatory response during insulitis. We also discuss how diet can affect the availability of fatty acids and disease development. Finally, we discuss development avenues that need further exploration. EXPERT OPINION Fatty acids, such as hydroxyl fatty acids, ω-3 fatty acids, and their downstream products, are druggable candidates that promote protective signaling. Inhibitors and antagonists of enzymes and receptors of arachidonic acid and free fatty acids, along with their derived metabolites, which cause pro-inflammatory and cytotoxic responses, have the potential to be developed as therapeutic targets also. Further, because diet is the main source of fatty acid intake in humans, balancing protective and pro-inflammatory/cytotoxic fatty acid levels through dietary therapy may have beneficial effects, delaying T1D progression. Therefore, therapeutic interventions targeting fatty acid signaling hold potential as avenues to treat T1D.
Collapse
Affiliation(s)
- Ivo Díaz Ludovico
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Soumyadeep Sarkar
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Emily Elliott
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Suvi M. Virtanen
- Health and Well-Being Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- Faculty of Social Sciences, Unit of Health Sciences, Tampere University, Tampere, Finland
- Tampere University Hospital, Research, Development and Innovation Center, Tampere, Finland
- Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Iris Erlund
- Department of Governmental Services, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology, and Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Raghavendra G. Mirmira
- Kovler Diabetes Center, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Thomas O. Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| |
Collapse
|
11
|
Ortiz-Placín C, Castillejo-Rufo A, Estarás M, González A. Membrane Lipid Derivatives: Roles of Arachidonic Acid and Its Metabolites in Pancreatic Physiology and Pathophysiology. Molecules 2023; 28:4316. [PMID: 37298790 PMCID: PMC10254454 DOI: 10.3390/molecules28114316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
One of the most important constituents of the cell membrane is arachidonic acid. Lipids forming part of the cellular membrane can be metabolized in a variety of cellular types of the body by a family of enzymes termed phospholipases: phospholipase A2, phospholipase C and phospholipase D. Phospholipase A2 is considered the most important enzyme type for the release of arachidonic acid. The latter is subsequently subjected to metabolization via different enzymes. Three enzymatic pathways, involving the enzymes cyclooxygenase, lipoxygenase and cytochrome P450, transform the lipid derivative into several bioactive compounds. Arachidonic acid itself plays a role as an intracellular signaling molecule. Additionally, its derivatives play critical roles in cell physiology and, moreover, are involved in the development of disease. Its metabolites comprise, predominantly, prostaglandins, thromboxanes, leukotrienes and hydroxyeicosatetraenoic acids. Their involvement in cellular responses leading to inflammation and/or cancer development is subject to intense study. This manuscript reviews the findings on the involvement of the membrane lipid derivative arachidonic acid and its metabolites in the development of pancreatitis, diabetes and/or pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | - Antonio González
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, 10003 Cáceres, Spain; (C.O.-P.); (A.C.-R.); (M.E.)
| |
Collapse
|
12
|
Functional Characterization of Transgenic Mice Overexpressing Human 15-Lipoxygenase-1 (ALOX15) under the Control of the aP2 Promoter. Int J Mol Sci 2023; 24:ijms24054815. [PMID: 36902243 PMCID: PMC10003068 DOI: 10.3390/ijms24054815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Arachidonic acid lipoxygenases (ALOX) have been implicated in the pathogenesis of inflammatory, hyperproliferative, neurodegenerative, and metabolic diseases, but the physiological function of ALOX15 still remains a matter of discussion. To contribute to this discussion, we created transgenic mice (aP2-ALOX15 mice) expressing human ALOX15 under the control of the aP2 (adipocyte fatty acid binding protein 2) promoter, which directs expression of the transgene to mesenchymal cells. Fluorescence in situ hybridization and whole-genome sequencing indicated transgene insertion into the E1-2 region of chromosome 2. The transgene was highly expressed in adipocytes, bone marrow cells, and peritoneal macrophages, and ex vivo activity assays proved the catalytic activity of the transgenic enzyme. LC-MS/MS-based plasma oxylipidome analyses of the aP2-ALOX15 mice suggested in vivo activity of the transgenic enzyme. The aP2-ALOX15 mice were viable, could reproduce normally, and did not show major phenotypic alterations when compared with wildtype control animals. However, they exhibited gender-specific differences with wildtype controls when their body-weight kinetics were evaluated during adolescence and early adulthood. The aP2-ALOX15 mice characterized here can now be used for gain-of-function studies evaluating the biological role of ALOX15 in adipose tissue and hematopoietic cells.
Collapse
|
13
|
Genotype-Phenotype Correlations in Human Diseases Caused by Mutations of LINC Complex-Associated Genes: A Systematic Review and Meta-Summary. Cells 2022; 11:cells11244065. [PMID: 36552829 PMCID: PMC9777268 DOI: 10.3390/cells11244065] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Mutations in genes encoding proteins associated with the linker of nucleoskeleton and cytoskeleton (LINC) complex within the nuclear envelope cause different diseases with varying phenotypes including skeletal muscle, cardiac, metabolic, or nervous system pathologies. There is some understanding of the structure of LINC complex-associated proteins and how they interact, but it is unclear how mutations in genes encoding them can cause the same disease, and different diseases with different phenotypes. Here, published mutations in LINC complex-associated proteins were systematically reviewed and analyzed to ascertain whether patterns exist between the genetic sequence variants and clinical phenotypes. This revealed LMNA is the only LINC complex-associated gene in which mutations commonly cause distinct conditions, and there are no clear genotype-phenotype correlations. Clusters of LMNA variants causing striated muscle disease are located in exons 1 and 6, and metabolic disease-associated LMNA variants are frequently found in the tail of lamin A/C. Additionally, exon 6 of the emerin gene, EMD, may be a mutation "hot-spot", and diseases related to SYNE1, encoding nesprin-1, are most often caused by nonsense type mutations. These results provide insight into the diverse roles of LINC-complex proteins in human disease and provide direction for future gene-targeted therapy development.
Collapse
|
14
|
Dong L, Wang H, Chen K, Li Y. Roles of hydroxyeicosatetraenoic acids in diabetes (HETEs and diabetes). Biomed Pharmacother 2022; 156:113981. [DOI: 10.1016/j.biopha.2022.113981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
|
15
|
Liu N, Liu Y, Dong D, Yu J, Yuan H. Effects of Inflammatory Factor Expression Regulated by 12/15 Lipoxygenase on Obesity-Related Nephropathy. Nutrients 2022; 14:nu14132743. [PMID: 35807921 PMCID: PMC9268756 DOI: 10.3390/nu14132743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/15/2022] [Accepted: 06/25/2022] [Indexed: 02/01/2023] Open
Abstract
Background: It has been demonstrated that 12/15-lipoxygenase (LO) contributes to insulin resistance by promoting beta cells’ exposure to inflammation. We investigate the mechanism by which 12/15-LO regulates the expression of inflammatory factors in obesity-related glomerular disease (ORG). Methods: Glomerular mesangial cells were treated with metabolite of 12/15-LO, and the expression of inflammatory factors was measured. Cell histones methylation in 12/15-LO related metabolic memory process were evaluated by chromatin immunoprecipitation (ChIP) assays. Wild-type (WT) and 12/15-LO knockout mice were fed a high-fat diet (HFD) to induce ORG. Results: 12(S)-HETE increased TNF-α, MCP-1, and IL-6 mRNA expression. Inhibition of 12/15-LO reduced the expression of inflammatory factors stimulated by PA or TNF-α. ChIP assays showed that 12(S)-HETE increased H3K4me modification in the TNF-α, IL-6, and MCP-1 gene promoters, and decreased H3K9me3 modification in the MCP-1 and IL-6 gene promoter. Urinary albumin excretion was greater in HFD-fed than in standard fat diet-fed mice, but both urinary protein and microalbumin amounts were lower in HFD-fed 12/15-LO knockout than in WT mice. The levels of TNF-α, IL-6, and MCP-1 in serum and renal cortex were higher in WT than in 12/15-LO knockout mice. Conclusions: 12/15-LO may regulate the expression of inflammatory factors in ORG by methylation of histones in the promoter regions of genes encoding inflammatory factors, sustaining the inflammatory phenotype of ORG.
Collapse
Affiliation(s)
- Nian Liu
- Department of Urology, First Hospital of Jilin University, Xin Min Street 1, Changchun 130021, China;
| | - Yang Liu
- Department of Nephrology, First Hospital of Jilin University, Xin Min Street 1, Changchun 130021, China; (Y.L.); (D.D.); (J.Y.)
| | - Dan Dong
- Department of Nephrology, First Hospital of Jilin University, Xin Min Street 1, Changchun 130021, China; (Y.L.); (D.D.); (J.Y.)
| | - Jinyu Yu
- Department of Nephrology, First Hospital of Jilin University, Xin Min Street 1, Changchun 130021, China; (Y.L.); (D.D.); (J.Y.)
| | - Hang Yuan
- Department of Nephrology, First Hospital of Jilin University, Xin Min Street 1, Changchun 130021, China; (Y.L.); (D.D.); (J.Y.)
- Correspondence: ; Tel.: +86-17604307906
| |
Collapse
|
16
|
Piñeros AR, Kulkarni A, Gao H, Orr KS, Glenn L, Huang F, Liu Y, Gannon M, Syed F, Wu W, Anderson CM, Evans-Molina C, McDuffie M, Nadler JL, Morris MA, Mirmira RG, Tersey SA. Proinflammatory signaling in islet β cells propagates invasion of pathogenic immune cells in autoimmune diabetes. Cell Rep 2022; 39:111011. [PMID: 35767947 PMCID: PMC9297711 DOI: 10.1016/j.celrep.2022.111011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 04/10/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes is a disorder of immune tolerance that leads to death of insulin-producing islet β cells. We hypothesize that inflammatory signaling within β cells promotes progression of autoimmunity within the islet microenvironment. To test this hypothesis, we deleted the proinflammatory gene encoding 12/15-lipoxygenase (Alox15) in β cells of non-obese diabetic mice at a pre-diabetic time point when islet inflammation is a feature. Deletion of Alox15 leads to preservation of β cell mass, reduces populations of infiltrating T cells, and protects against spontaneous autoimmune diabetes in both sexes. Mice lacking Alox15 in β cells exhibit an increase in a population of β cells expressing the gene encoding the protein programmed death ligand 1 (PD-L1), which engages receptors on immune cells to suppress autoimmunity. Delivery of a monoclonal antibody against PD-L1 recovers the diabetes phenotype in knockout animals. Our results support the contention that inflammatory signaling in β cells promotes autoimmunity during type 1 diabetes progression.
Collapse
Affiliation(s)
- Annie R Piñeros
- Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Abhishek Kulkarni
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kara S Orr
- Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lindsey Glenn
- Department of Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Fei Huang
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maureen Gannon
- Department of Medicine, Vanderbilt University and Department of Veterans Affairs, Tennessee Valley Authority, Nashville, TN, USA
| | - Farooq Syed
- Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wenting Wu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cara M Anderson
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Carmella Evans-Molina
- Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Marcia McDuffie
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Jerry L Nadler
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Margaret A Morris
- Department of Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Raghavendra G Mirmira
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA.
| | - Sarah A Tersey
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
17
|
Isse FA, El-Sherbeni AA, El-Kadi AOS. The multifaceted role of cytochrome P450-Derived arachidonic acid metabolites in diabetes and diabetic cardiomyopathy. Drug Metab Rev 2022; 54:141-160. [PMID: 35306928 DOI: 10.1080/03602532.2022.2051045] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding lipid metabolism is a critical key to understanding the pathogenesis of Diabetes Mellitus (DM). It is known that 60-90% of DM patients are obese or used to be obese. The incidence of obesity is rising owing to the modern sedentary lifestyle that leads to insulin resistance and increased levels of free fatty acids, predisposing tissues to utilize more lipids with less glucose uptake. However, the exact mechanism is not yet fully elucidated. Diabetic cardiomyopathy seems to be associated with these alterations in lipid metabolism. Arachidonic acid (AA) is an important fatty acid that is metabolized to several bioactive compounds by cyclooxygenases, lipoxygenases, and the more recently discovered, cytochrome P450 (P450) enzymes. P450 metabolizes AA to either epoxy-AA (EETs) or hydroxy-AA (HETEs). Studies showed that EETs could have cardioprotective effects and beneficial effects in reversing abnormalities in glucose and insulin homeostasis. Conversely, HETEs, most importantly 12-HETE and 20-HETE, were found to interfere with normal glucose and insulin homeostasis and thus, might be involved in diabetic cardiomyopathy. In this review, we highlight the role of P450-derived AA metabolites in the context of DM and diabetic cardiomyopathy and their potential use as a target for developing new treatments for DM and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Fadumo Ahmed Isse
- Departmet of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Ahmed A El-Sherbeni
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Ayman O S El-Kadi
- Departmet of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
18
|
Chen S, Zou H. Key Role of 12-Lipoxygenase and Its Metabolite 12-Hydroxyeicosatetraenoic Acid (12-HETE) in Diabetic Retinopathy. Curr Eye Res 2022; 47:329-335. [PMID: 35129022 DOI: 10.1080/02713683.2021.1995003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE Abnormal lipid metabolism has been proved to be implicated in the complex pathogenesis of diabetic retinopathy (DR). 12-lipoxygenase (12-LOX) is a member of lipoxygenase family responsible for the oxygenation of cellular polyunsaturated fatty acids to produce lipid mediators which modulate cell inflammation. This review explores the role of 12-lipoxygenase and its products in the pathogenesis of DR. METHODS A comprehensive medical literature search was conducted on PubMed till September 2021. RESULTS Emerging evidence has demonstrated that 12-LOX and its main product 12- hydroxyeicosatetraenoic acid (12-HETE) activate retinal cells, especially retinal vascular endothelial cells, through the activation of NADPH oxidase and the subsequent generation of reactive oxygen species (ROS), mediating multiple pathological changes during DR. Genetic deletion or pharmacological inhibition models of 12-LOX in mice show protection from DR. CONCLUSION 12-LOX and its product 12-HETE take important part in DR pathogenesis and show their potential as future therapeutic targets for DR. Further studies are needed on the specific mechanism including 12-LOX pathway related molecules, 12-HETE receptors and downstream signaling pathways.
Collapse
Affiliation(s)
- Shuli Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Haidong Zou
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Eye Disease Prevention and Treatment Center, Shanghai Eye Hospital, Shanghai, China.,Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
19
|
Choudhary R, Kumar M, Katyal A. 12/15-Lipoxygenase debilitates mitochondrial health in intermittent hypobaric hypoxia induced neuronal damage: An in vivo study. Redox Biol 2022; 49:102228. [PMID: 34979449 PMCID: PMC8728585 DOI: 10.1016/j.redox.2021.102228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 12/31/2022] Open
Abstract
High altitude cerebral edema does not fall in routine definition of hypoxia and requires alternative therapeutic strategies. 12/15-Lipoxygenase (12/15 LOX), a key proinflammatory lipid peroxidative enzyme which site specifically inserts into cellular and subcellular membranes and plays an instrumental role in hypobaric hypoxia induced neuropathogenesis. Mitochondria, the master regulator organelles for oxygen consumption and ATP generation are sensitive to intracellular oxygen perturbations and are associated with activation of apoptosis based cell death cascades that seal the fate of the cell. The mechanistic involvement of 12/15 LOX in mitochondria mediated cell death in brain microenvironment during hypobaric hypoxia conditions can be an interesting preposition. In the present study, we have investigated underlying involvement of 12/15 LOX in hypobaric hypoxia (HH) induced disturbance in mitochondrial integrity and its relation with neuronal apoptosis. Male Balb/c mice subjected to simulated HH condition for three consecutive days showed robust increase in intra-hippocampal 12(S)HETE (12/15 LOX metabolite), which was significantly reduced following baicalein (12/15 LOX Inhibitor) treatment. The elevated level of 12(S)HETE following hypobaric hypoxia condition correlated with simultaneous increase in expression of 12/15 LOX in neurons and microglia lining the hippocampal CA3 region. Further, 12/15 LOX gets embedded onto the periphery of mitochondria following HH and a strong correlation has been observed with loss of mitochondrial integrity as evident from increased cytochrome-c in the cytosolic compartment and a subsequent upregulated activity of Caspase-3 and Caspase-9 as well as Bax/Bcl-2 expression ratio. The observed effects seen under HH were reversed upon treatment with baicalein suggesting a critical role of 12/15 LOX in HH induced mitochondrial damage Further, the hypobaric hypoxia-mediated increase in hippocampal pAKT and pmTOR protein expression were significantly ameliorated following 12/15 LOX inhibition, suggesting a mitochondrial involvement. We hereby demonstrate the contribution of 12/15 LOX in disorienting mitochondrial integrity with subsequent release of cytochrome-c in cytosol which drives the neuronal cells to intrinsic mode of cell death during hypobaric hypoxia. The protective role of baicalein by inhibition of 12/15 LOX dependent neuronal cell death and preservation of mitochondrial integrity suggests it to be a plausible therapeutic target in CNS related disorders.
Collapse
Affiliation(s)
- Richa Choudhary
- Dr. B.R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| | - Mukesh Kumar
- Dr. B.R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| | - Anju Katyal
- Dr. B.R Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India.
| |
Collapse
|
20
|
Kriska T, Herrnreiter A, Pfister SL, Adebesin A, Falck JR, Campbell WB. Macrophage 12(S)-HETE Enhances Angiotensin II-Induced Contraction by a BLT2 (Leukotriene B 4 Type-2 Receptor) and TP (Thromboxane Receptor)-Mediated Mechanism in Murine Arteries. Hypertension 2022; 79:104-114. [PMID: 34784723 PMCID: PMC8849474 DOI: 10.1161/hypertensionaha.121.17824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
12/15-LO (12/15-lipoxygenase), encoded by Alox15 gene, metabolizes arachidonic acid to 12(S)-HETE (12-hydroxyeicosatetraenoic acid). Macrophages are the major source of 12/15-LO among immune cells, and 12/15-LO plays a crucial role in development of hypertension. Global Alox15- or macrophage-deficient mice are resistant to Ang II (angiotensin II)-induced hypertension. This study tests the hypothesis that macrophage 12(S)-HETE contributes to Ang II-mediated arterial constriction and thus to development of Ang II-induced hypertension. Ang II constricted isolated abdominal aortic and mesenteric arterial rings. 12(S)-HETE (100 nmol/L) alone was without effect; however, it significantly enhanced Ang II-induced constriction. The presence of wild-type macrophages also enhanced the Ang II-induced constriction, while Alox15-/- macrophages did not. Using this model, pretreatment of aortic rings with inhibitors, receptor agonists/antagonists, or removal of the endothelium, systematically uncovered an endothelium-mediated, Ang II receptor-2-mediated and superoxide-mediated enhancing effect of 12(S)-HETE on Ang II constrictions. The role of superoxide was confirmed using aortas from p47phox-/- mice where 12(S)-HETE failed to enhance constriction to Ang II. In cultured arterial endothelial cells, 12(S)-HETE increased the production of superoxide, and 12(S)-HETE or Ang II increased the production of an isothromboxane-like metabolite. A TP (thromboxane receptor) antagonist inhibited 12(S)-HETE enhancement of Ang II constriction. Both Ang II-induced hypertension and the enhancing effect of 12(S)-HETE on Ang II contractions were eliminated by a BLT2 (leukotriene B4 receptor-2) antagonist. These results outline a mechanism where the macrophage 12/15-LO pathway enhances the action of Ang II. 12(S)-HETE, acting on the BLT2, contributes to the hypertensive action of Ang II in part by promoting endothelial synthesis of a superoxide-derived TP agonist.
Collapse
Affiliation(s)
- Tamas Kriska
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee (T.K., A.H., S.L.P., W.B.C.)
| | - Anja Herrnreiter
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee (T.K., A.H., S.L.P., W.B.C.)
| | - Sandra L Pfister
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee (T.K., A.H., S.L.P., W.B.C.)
| | - Adeniyi Adebesin
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (A.A., J.R.F.)
| | - John R Falck
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas (A.A., J.R.F.)
| | - William B Campbell
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee (T.K., A.H., S.L.P., W.B.C.)
| |
Collapse
|
21
|
Zhang XJ, Ji YX, Cheng X, Cheng Y, Yang H, Wang J, Zhao LP, Huang YP, Sun D, Xiang H, Shen LJ, Li PL, Ma JP, Tian RF, Yang J, Yao X, Xu H, Liao R, Xiao L, Zhang P, Zhang X, Zhao GN, Wang X, Hu ML, Tian S, Wan J, Cai J, Ma X, Xu Q, Wang Y, Touyz RM, Liu PP, Loomba R, She ZG, Li H. A small molecule targeting ALOX12-ACC1 ameliorates nonalcoholic steatohepatitis in mice and macaques. Sci Transl Med 2021; 13:eabg8116. [PMID: 34910548 DOI: 10.1126/scitranslmed.abg8116] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) is a progressive liver disease and has become a leading indication for liver transplantation in the United States. The development of effective therapies for NASH is a major unmet need. Here, we identified a small molecule, IMA-1, that can treat NASH by interrupting the arachidonate 12-lipoxygenase (ALOX12)–acetyl-CoA carboxylase 1 (ACC1) interaction. IMA-1 markedly blocked diet-induced NASH progression in both male mice and Cynomolgus macaque therapeutic models. The anti-NASH efficacy of IMA-1 was comparable to ACC inhibitor in both species. Protein docking simulations and following functional experiments suggested that the anti-NASH effects of IMA-1 were largely dependent on its direct binding to a pocket in ALOX12 proximal to its ACC1-interacting surface instead of inhibiting ALOX12 lipoxygenase activity. IMA-1 treatment did not elicit hyperlipidemia, a known side effect of direct inhibition of ACC enzymatic activity, in both mice and macaques. These findings provide proof of concept across multiple species for the use of small molecule–based therapies for NASH.
Collapse
Affiliation(s)
- Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yan-Xiao Ji
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xu Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yanjie Cheng
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hailong Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Junyong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Ling-Ping Zhao
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yong-Ping Huang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Dating Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hui Xiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Li-Jun Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Peng-Long Li
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Jun-Peng Ma
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Rui-Feng Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Juan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xinxin Yao
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Rufang Liao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Li Xiao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Peng Zhang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xin Zhang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Guang-Nian Zhao
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Man-Li Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Juan Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19004, USA
| | - Qingbo Xu
- Centre for Clinic Pharmacology, The William Harvey Research Institute, Queen Mary University of London, London SE5 9NU, UK
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rhian M Touyz
- British Heart Foundation Chair in Cardiovascular Medicine, and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Peter P Liu
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Epidemiology, University of California, San Diego, San Diego, CA 92093, USA
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
22
|
Zhang XJ, She ZG, Wang J, Sun D, Shen LJ, Xiang H, Cheng X, Ji YX, Huang YP, Li PL, Yang X, Cheng Y, Ma JP, Wang HP, Hu Y, Hu F, Tian S, Tian H, Zhang P, Zhao GN, Wang L, Hu ML, Yang Q, Zhu LH, Cai J, Yang J, Zhang X, Ma X, Xu Q, Touyz RM, Liu PP, Loomba R, Wang Y, Li H. Multiple omics study identifies an interspecies conserved driver for nonalcoholic steatohepatitis. Sci Transl Med 2021; 13:eabg8117. [PMID: 34910546 DOI: 10.1126/scitranslmed.abg8117] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lipotoxicity is a recognized pathological trigger and accelerator of nonalcoholic steatohepatitis (NASH). However, the molecular basis of lipotoxicity-induced NASH remains elusive. Here, we systematically mapped the changes in hepatic transcriptomic landscapes in response to lipotoxic insults across multiple species. Conserved and robust activation of the arachidonic acid pathway, in particular the arachidonate 12-lipoxygenase (ALOX12) gene, was closely correlated with NASH severity in humans, macaques with spontaneously developed NASH, as well as swine and mouse dietary NASH models. Using gain- and loss-of-function studies, we found that ALOX12 markedly exacerbated NASH in both mice and Bama pig models. ALOX12 was shown to induce NASH by directly targeting acetyl-CoA carboxylase 1 (ACC1) via a lysosomal degradation mechanism. Overall, our findings reveal a key molecular driver of NASH pathogenesis and suggest that ALOX12-ACC1 interaction may be a therapeutic target in NASH.
Collapse
Affiliation(s)
- Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Junyong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Dating Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Li-Jun Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hui Xiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xu Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yan-Xiao Ji
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yong-Ping Huang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Peng-Long Li
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xia Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yanjie Cheng
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jun-Peng Ma
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Hai-Ping Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yufeng Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Fengjiao Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Han Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Peng Zhang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Guang-Nian Zhao
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Lin Wang
- Department of Hepatic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Man-Li Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Qin Yang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Li-Hua Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Juan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xin Zhang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19004, USA
| | - Qingbo Xu
- Centre for Clinic Pharmacology, The William Harvey Research Institute, Queen Mary University of London, London SE5 9NU, UK
| | - Rhian M Touyz
- British Heart Foundation Chair in Cardiovascular Medicine, and Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Peter P Liu
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Epidemiology, University of California, San Diego, San Diego, CA 92093, USA
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Model Animal of Wuhan University, Wuhan 430071, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
23
|
Otto M, Brabenec L, Müller M, Kintrup S, Hellenthal KEM, Holtmeier R, Steinbuch SC, Karsten OS, Pryvalov H, Rossaint J, Gross ER, Wagner NM. Development of heart failure with preserved ejection fraction in type 2 diabetic mice is ameliorated by preserving vascular function. Life Sci 2021; 284:119925. [PMID: 34480933 PMCID: PMC8484044 DOI: 10.1016/j.lfs.2021.119925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 08/13/2021] [Accepted: 08/21/2021] [Indexed: 11/26/2022]
Abstract
AIMS Heart failure with preserved ejection fraction (HFpEF) is associated with endothelial dysfunction and is frequent in people with type 2 diabetes mellitus. In diabetic patients, increased levels of the eicosanoid 12-hydroxyeicosatetraenoic acid (12-HETE) are linked to vascular dysfunction. Here, we aimed to identify the importance of 12-HETE in type 2 diabetic patients exhibiting diastolic dysfunction, and mice exhibiting HFpEF and whether targeting 12-HETE is a means to ameliorate HFpEF progression by improving vascular function in diabetes. MATERIAL AND METHODS Subjects with diagnosed type 2 diabetes mellitus and reported diastolic dysfunction or healthy controls were recruited and 12(S)-HETE levels determined by ELISA. 12(S)-HETE levels were determined in type 2 diabetic, leptin receptor deficient mice (LepRdb/db) and HFpEF verified by echocardiography. Mitochondrial function, endothelial function and capillary density were assessed using Seahorse technique, pressure myography and immunohistochemistry in LepRdb/db or non-diabetic littermate controls. 12/15Lo generation was inhibited using ML351 and 12(S)-HETE action by using the V1-cal peptide. KEY FINDINGS Endothelium-dependent vasodilation and mitochondrial functional capacity both improved in response to either application of ML351 or the V1-cal peptide. Correlating to improved vascular function, mice treated with either pharmacological agent exhibited improved diastolic filling and left ventricular relaxation that correlated with increased myocardial capillary density. SIGNIFICANCE Our results suggest that 12-HETE may serve as a biomarker indicating endothelial dysfunction and the resulting cardiovascular consequences such as HFpEF in type 2 diabetic patients. Antagonizing 12-HETE is a potent means to causally control HFpEF development and progression in type 2 diabetes by preserving vascular function.
Collapse
Affiliation(s)
- Mandy Otto
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Laura Brabenec
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Melanie Müller
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Sebastian Kintrup
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Katharina E M Hellenthal
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Richard Holtmeier
- Institute of Clinical Radiology, University Hospital Muenster, Muenster, Germany
| | - Sophie Charlotte Steinbuch
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Ole Sönken Karsten
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Heorhii Pryvalov
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Eric R Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Nana-Maria Wagner
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany.
| |
Collapse
|
24
|
Shichiri M, Ishida N, Aoki Y, Koike T, Hagihara Y. Stress-activated leukocyte 12/15-lipoxygenase metabolite enhances struggle behaviour and tocotrienols relieve stress-induced behaviour alteration. Free Radic Biol Med 2021; 175:171-183. [PMID: 34474105 DOI: 10.1016/j.freeradbiomed.2021.08.236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/05/2021] [Accepted: 08/11/2021] [Indexed: 11/22/2022]
Abstract
Stress induces emotional arousal causing anxiety, irritability, exaggerated startle behaviour, and hypervigilance observed in patients with trauma and stress-related mental disorders, including acute stress disorder and post-traumatic stress disorder. Central norepinephrine release promotes stress-induced emotional arousal. However, the regulator of emotional arousal remains unknown. Here, we show that the arachidonate-derived metabolite produced by stress-activated leukocyte 12/15-lipoxygenase is remarkably elevated in the plasma and upregulates the central norepinephrine release, resulting in the enhancement of the struggle behaviour (= escape behaviour) in the tail suspension test. Struggle behaviour is mimicking a symptom of emotional arousal. This stress-induced struggle behaviour was absent in 12/15-lipoxygenase deficient mice; however, intravenous administration of a 12/15-lipoxygenase metabolite to these mice after stress exposure rekindled the struggle behaviour. Furthermore, tocotrienols and geranylgeraniol reduced stress-induced 12/15-lipoxygenase metabolite production and suppressed the struggle behaviour. Our findings indicate that arachidonate-derived 12/15-lipoxygenase metabolite is involved in the regulation of stress-enhanced central norepinephrine release and struggle behaviour. In addition, we propose 12/15-lipoxygenase as a potential therapeutic target for the treatment of emotional arousal observed in stress-related mental disorders.
Collapse
Affiliation(s)
- Mototada Shichiri
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan; DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), 1-1-1 Higashi, Tsukuba-shi, Ibaraki, 305-8562, Japan.
| | - Noriko Ishida
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan
| | - Yoshinori Aoki
- Healthcare Solutions Unit, Life Solutions Sector, Amenity Life Division, Advanced Solutions Domain, Mitsubishi Chemical Corporation, 1-1-1, Marunouchi, Chiyoda-ku, Tokyo, 100-8251, Japan
| | - Taisuke Koike
- Strategy Department, Advanced Solutions Planning Division, Advanced Solutions Domain, Mitsubishi Chemical Corporation, 1-1-1, Marunouchi, Chiyoda-ku, Tokyo, 100-8251, Japan
| | - Yoshihisa Hagihara
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan
| |
Collapse
|
25
|
Zhang XJ, Liu X, Hu M, Zhao GJ, Sun D, Cheng X, Xiang H, Huang YP, Tian RF, Shen LJ, Ma JP, Wang HP, Tian S, Gan S, Xu H, Liao R, Zou T, Ji YX, Zhang P, Cai J, Wang ZV, Meng G, Xu Q, Wang Y, Ma XL, Liu PP, Huang Z, Zhu L, She ZG, Zhang X, Bai L, Yang H, Lu Z, Li H. Pharmacological inhibition of arachidonate 12-lipoxygenase ameliorates myocardial ischemia-reperfusion injury in multiple species. Cell Metab 2021; 33:2059-2075.e10. [PMID: 34536344 DOI: 10.1016/j.cmet.2021.08.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/01/2020] [Accepted: 08/25/2021] [Indexed: 12/18/2022]
Abstract
Myocardial ischemia-reperfusion (MIR) injury is a major cause of adverse outcomes of revascularization after myocardial infarction. To identify the fundamental regulator of reperfusion injury, we performed metabolomics profiling in plasma of individuals before and after revascularization and identified a marked accumulation of arachidonate 12-lipoxygenase (ALOX12)-dependent 12-HETE following revascularization. The potent induction of 12-HETE proceeded by reperfusion was conserved in post-MIR in mice, pigs, and monkeys. While genetic inhibition of Alox12 protected mouse hearts from reperfusion injury and remodeling, Alox12 overexpression exacerbated MIR injury. Remarkably, pharmacological inhibition of ALOX12 significantly reduced cardiac injury in mice, pigs, and monkeys. Unexpectedly, ALOX12 promotes cardiomyocyte injury beyond its enzymatic activity and production of 12-HETE but also by its suppression of AMPK activity via a direct interaction with its upstream kinase TAK1. Taken together, our study demonstrates that ALOX12 is a novel AMPK upstream regulator in the post-MIR heart and that it represents a conserved therapeutic target for the treatment of myocardial reperfusion injury.
Collapse
Affiliation(s)
- Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xiaolan Liu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Manli Hu
- Institute of Model Animal of Wuhan University, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Guo-Jun Zhao
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Dating Sun
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xu Cheng
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hui Xiang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yong-Ping Huang
- Institute of Model Animal of Wuhan University, Wuhan 430071, China; College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Rui-Feng Tian
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Li-Jun Shen
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Jun-Peng Ma
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Hai-Ping Wang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Shanyu Gan
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Rufang Liao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Toujun Zou
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Yan-Xiao Ji
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Peng Zhang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Jingjing Cai
- Department of Cardiology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guannan Meng
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China
| | - Qingbo Xu
- Centre for Clinic Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xin-Liang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA 19004, USA
| | - Peter P Liu
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Zan Huang
- College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Lihua Zhu
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China
| | - Xin Zhang
- Gannan Institute of Translational Medicine, Ganzhou 341000, China
| | - Lan Bai
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China.
| | - Hailong Yang
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China.
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430060, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital, School of Basic Medical Science, Wuhan University, Wuhan 430071, China; Institute of Model Animal of Wuhan University, Wuhan 430071, China; Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
26
|
Tsai WC, Aleem AM, Tena J, Rivera-Velazquez M, Brah HS, Tripathi S, D'silva M, Nadler JL, Kalyanaraman C, Jacobson MP, Holman T. Docking and mutagenesis studies lead to improved inhibitor development of ML355 for human platelet 12-lipoxygenase. Bioorg Med Chem 2021; 46:116347. [PMID: 34507163 DOI: 10.1016/j.bmc.2021.116347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/14/2021] [Accepted: 07/30/2021] [Indexed: 11/16/2022]
Abstract
Human platelet 12-(S)-Lipoxygenase (12-LOX) is a fatty acid metabolizing oxygenase that plays an important role in platelet activation and cardiometabolic disease. ML355 is a specific 12-LOX inhibitor that has been shown to decrease thrombosis without prolonging hemostasis and protect human pancreatic islets from inflammatory injury. It has an amenable drug-like scaffold with nM potency and encouraging ADME and PK profiles, but its binding mode to the active site of 12-LOX remains unclear. In the current work, we combined computational modeling and experimental mutagenesis to propose a model in which ML355 conforms to the "U" shape of the 12-LOX active site, with the phenyl linker region wrapping around L407. The benzothiazole of ML355 extends into the bottom of the active site cavity, pointing towards residues A417 and V418. However, reducing the active site depth alone did not affect ML355 potency. In order to lower the potency of ML355, the cavity needed to be reduced in both length and width. In addition, H596 appears to position ML355 in the active site through an interaction with the 2-methoxy phenol moiety of ML355. Combined, this binding model suggested that the benzothiazole of ML355 could be enlarged. Therefore, a naphthyl-benzothiazole derivative of ML355, Lox12Slug001, was synthesized and shown to have 7.2-fold greater potency than ML355. This greater potency is proposed to be due to additional van der Waals interactions and pi-pi stacking with F414 and F352. Lox12Slug001 was also shown to be highly selective against 12-LOX relative to the other LOX isozymes and more importantly, it showed activity in rescuing human islets exposed to inflammatory cytokines with comparable potency to ML355. Further studies are currently being pursued to derivatize ML355 in order to optimize the additional space in the active site, while maintaining acceptable drug-like properties.
Collapse
Affiliation(s)
- Wan-Chen Tsai
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, United States
| | - Ansari M Aleem
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, United States
| | - Jennyfer Tena
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, United States
| | - Mirella Rivera-Velazquez
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, United States
| | - Harman Singh Brah
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA 94143, United States
| | - Sarvind Tripathi
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, United States
| | - Melinee D'silva
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Jerry L Nadler
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | - Chakrapani Kalyanaraman
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA 94143, United States
| | - Matthew P Jacobson
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California San Francisco, San Francisco, CA 94143, United States
| | - Theodore Holman
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA 95064, United States.
| |
Collapse
|
27
|
Yao D, Qiao F, Song C, Lv Y. Matrix stiffness regulates bone repair by modulating 12-lipoxygenase-mediated early inflammation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112359. [PMID: 34474906 DOI: 10.1016/j.msec.2021.112359] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/04/2021] [Accepted: 08/04/2021] [Indexed: 12/21/2022]
Abstract
Lipid metabolism in macrophages has been increasingly emphasized in exerting an anti-inflammatory effect and accelerating fracture healing. 12-lipoxygenase (12-LOX) is expressed in several cell types, including macrophages, and oxidizes polyunsaturated fatty acids (PUFAs) to generate both pro- and anti-inflammatory lipid mediators, of which the n-3 PUFAs play an important part in tissue homeostasis/fibrosis. Although mechanical factor regulates the lipid metabolic axis of inflammatory cells, specifically matrix stiffness influences macrophages metabolic responses, little is known about how matrix stiffness affects the 12-LOX-mediated early inflammation in bone repair. In the present study, demineralized bone matrix (DBM) scaffolds with different matrix stiffness were constructed by controlling the duration of decalcification (0 h (control), 1 h (high), 12 h (medium), and 5 d (low)) to repair the defected rat skull. The expression of inflammatory cytokines and macrophages polarization were analyzed. The lipid metabolites and lipid mediators' biosynthesis by matrix stiffness-regulated were further detected. The results showed that the low matrix stiffness could polarize macrophages into an anti-inflammatory phenotype, promote the expression of anti-inflammatory cytokines and specialized pro-resolving lipid mediators (SPMs) biosynthesis beneficial for the osteogenesis of mesenchymal stem cells (MSCs). After treated with ML355, the expression of anti-inflammatory cytokines/proteins and SPMs biosynthesis in macrophages cultured on low-matrix stiffness scaffolds were repressed, and there were almost no statistical differences among all groups. Findings from this study support that matrix stiffness regulates bone repair by modulating 12-LOX-mediated early inflammation, which suggest a direct mechanical impact of matrix stiffness on macrophages lipid metabolism and provide a new insight into the clinical application of SPMs for bone regeneration.
Collapse
Affiliation(s)
- Dongdong Yao
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Fangyu Qiao
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Chenchen Song
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Yonggang Lv
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China.
| |
Collapse
|
28
|
Kulkarni A, Pineros AR, Walsh MA, Casimiro I, Ibrahim S, Hernandez-Perez M, Orr KS, Glenn L, Nadler JL, Morris MA, Tersey SA, Mirmira RG, Anderson RM. 12-Lipoxygenase governs the innate immune pathogenesis of islet inflammation and autoimmune diabetes. JCI Insight 2021; 6:e147812. [PMID: 34128835 PMCID: PMC8410073 DOI: 10.1172/jci.insight.147812] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Macrophages and related myeloid cells are innate immune cells that participate in the early islet inflammation of type 1 diabetes (T1D). The enzyme 12-lipoxygenase (12-LOX) catalyzes the formation of proinflammatory eicosanoids, but its role and mechanisms in myeloid cells in the pathogenesis of islet inflammation have not been elucidated. Leveraging a model of islet inflammation in zebrafish, we show here that macrophages contribute significantly to the loss of β cells and the subsequent development of hyperglycemia. The depletion or inhibition of 12-LOX in this model resulted in reduced macrophage infiltration into islets and the preservation of β cell mass. In NOD mice, the deletion of the gene encoding 12-LOX in the myeloid lineage resulted in reduced insulitis with reductions in proinflammatory macrophages, a suppressed T cell response, preserved β cell mass, and almost complete protection from the development of T1D. 12-LOX depletion caused a defect in myeloid cell migration, a function required for immune surveillance and tissue injury responses. This effect on migration resulted from the loss of the chemokine receptor CXCR3. Transgenic expression of the gene encoding CXCR3 rescued the migratory defect in zebrafish 12-LOX morphants. Taken together, our results reveal a formative role for innate immune cells in the early pathogenesis of T1D and identify 12-LOX as an enzyme required to promote their prodiabetogenic phenotype in the context of autoimmunity.
Collapse
Affiliation(s)
- Abhishek Kulkarni
- Center for Diabetes and Metabolic Diseases and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Annie R Pineros
- Center for Diabetes and Metabolic Diseases and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Melissa A Walsh
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Isabel Casimiro
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Sara Ibrahim
- Center for Diabetes and Metabolic Diseases and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Marimar Hernandez-Perez
- Center for Diabetes and Metabolic Diseases and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kara S Orr
- Center for Diabetes and Metabolic Diseases and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lindsey Glenn
- Department of Medicine, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Jerry L Nadler
- Department of Medicine, New York Medical College, Valhalla, New York, USA
| | - Margaret A Morris
- Department of Medicine, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Sarah A Tersey
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Raghavendra G Mirmira
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Ryan M Anderson
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
29
|
Wong XK, Yeong KY. A Patent Review on the Current Developments of Benzoxazoles in Drug Discovery. ChemMedChem 2021; 16:3237-3262. [PMID: 34289258 DOI: 10.1002/cmdc.202100370] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/11/2021] [Indexed: 12/11/2022]
Abstract
The benzoxazole moiety is widely found in various natural compounds, which are often found to be biologically active. Due to its versatile biological properties, benzoxazole has been incorporated as an essential pharmacophore and substructure in many medicinal compounds. In the past years, numerous benzoxazole derivatives have been synthesised and evaluated for their biological potential. The wide range in therapeutic potential of benzoxazole derivatives is related to the favourable interactions of the benzoxazole moiety with different protein targets. Herein we review the biological activities of benzoxazole derivatives patented within the past six years. Using the Lens database, granted patents issued from 2015 to 2020 were retrieved. The patented benzoxazole derivatives demonstrated excellent activity against various protein targets and diseases, with some reaching clinical trial stage. Pharmacological and medicinal aspects of patented benzoxazole derivatives are discussed. The recent development and drawbacks are also reviewed.
Collapse
Affiliation(s)
- Xi Khai Wong
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Keng Yoon Yeong
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| |
Collapse
|
30
|
Pascale JV, Lucchesi PA, Garcia V. Unraveling the Role of 12- and 20- HETE in Cardiac Pathophysiology: G-Protein-Coupled Receptors, Pharmacological Inhibitors, and Transgenic Approaches. J Cardiovasc Pharmacol 2021; 77:707-717. [PMID: 34016841 PMCID: PMC8523029 DOI: 10.1097/fjc.0000000000001013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/03/2021] [Indexed: 12/17/2022]
Abstract
ABSTRACT Arachidonic acid-derived lipid mediators play crucial roles in the development and progression of cardiovascular diseases. Eicosanoid metabolites generated by lipoxygenases and cytochrome P450 enzymes produce several classes of molecules, including the epoxyeicosatrienoic acid (EET) and hydroxyeicosatetraenoic acids (HETE) family of bioactive lipids. In general, the cardioprotective effects of EETs have been documented across a number of cardiac diseases. In contrast, members of the HETE family have been shown to contribute to the pathogenesis of ischemic cardiac disease, maladaptive cardiac hypertrophy, and heart failure. The net effect of 12(S)- and 20-HETE depends upon the relative amounts generated, ratio of HETEs:EETs produced, timing of synthesis, as well as cellular and subcellular mechanisms activated by each respective metabolite. HETEs are synthesized by and affect multiple cell types within the myocardium. Moreover, cytochrome P450-derived and lipoxygenase- derived metabolites have been shown to directly influence cardiac myocyte growth and the regulation of cardiac fibroblasts. The mechanistic data uncovered thus far have employed the use of enzyme inhibitors, HETE antagonists, and the genetic manipulation of lipid-producing enzymes and their respective receptors, all of which influence a complex network of outcomes that complicate data interpretation. This review will summarize and integrate recent findings on the role of 12(S)-/20-HETE in cardiac diseases.
Collapse
Affiliation(s)
| | | | - Victor Garcia
- Department of Pharmacology, New York Medical College, Valhalla, NY
| |
Collapse
|
31
|
Kulkarni A, Nadler JL, Mirmira RG, Casimiro I. Regulation of Tissue Inflammation by 12-Lipoxygenases. Biomolecules 2021; 11:717. [PMID: 34064822 PMCID: PMC8150372 DOI: 10.3390/biom11050717] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/03/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
Lipoxygenases (LOXs) are lipid metabolizing enzymes that catalyze the di-oxygenation of polyunsaturated fatty acids to generate active eicosanoid products. 12-lipoxygenases (12-LOXs) primarily oxygenate the 12th carbon of its substrates. Many studies have demonstrated that 12-LOXs and their eicosanoid metabolite 12-hydroxyeicosatetraenoate (12-HETE), have significant pathological implications in inflammatory diseases. Increased level of 12-LOX activity promotes stress (both oxidative and endoplasmic reticulum)-mediated inflammation, leading to damage in these tissues. 12-LOXs are also associated with enhanced cellular migration of immune cells-a characteristic of several metabolic and autoimmune disorders. Genetic depletion or pharmacological inhibition of the enzyme in animal models of various diseases has shown to be protective against disease development and/or progression in animal models in the setting of diabetes, pulmonary, cardiovascular, and metabolic disease, suggesting a translational potential of targeting the enzyme for the treatment of several disorders. In this article, we review the role of 12-LOXs in the pathogenesis of several diseases in which chronic inflammation plays an underlying role.
Collapse
Affiliation(s)
- Abhishek Kulkarni
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA;
| | - Jerry L. Nadler
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, NY 10595, USA;
| | | | - Isabel Casimiro
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA;
| |
Collapse
|
32
|
Rhee SY, Jung ES, Suh DH, Jeong SJ, Kim K, Chon S, Yu SY, Woo JT, Lee CH. Plasma amino acids and oxylipins as potential multi-biomarkers for predicting diabetic macular edema. Sci Rep 2021; 11:9727. [PMID: 33958610 PMCID: PMC8102569 DOI: 10.1038/s41598-021-88104-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/06/2021] [Indexed: 11/09/2022] Open
Abstract
To investigate the pathophysiologic characteristics of diabetic complications, we identified differences in plasma metabolites in subjects with type 2 diabetes (T2DM) with or without diabetic macular edema (DME) and a disease duration > 15 years. An cohort of older T2DM patients with prolonged disease duration was established, and clinical information and biospecimens were collected following the guidelines of the National Biobank of Korea. DME phenotypes were identified by ophthalmologic specialists. For metabolomics studies, propensity matched case and control samples were selected. To discover multi-biomarkers in plasma, non-targeted metabolite profiling and oxylipin profiling in the discovery cohort were validated in an extended cohort. From metabolomic studies, 5 amino acids (asparagine, aspartic acid, glutamic acid, cysteine, and lysine), 2 organic compounds (citric acid and uric acid) and 4 oxylipins (12-oxoETE, 15-oxoETE, 9-oxoODE, 20-carboxy leukotriene B4) were identified as candidate multi-biomarkers which can guide DME diagnosis among non-DME subjects. Receiver operating characteristic curves revealed high diagnostic value of the combined 5 amino acids and 2 organic compounds (AUC = 0.918), and of the 4 combined oxylipins (AUC = 0.957). Our study suggests that multi-biomarkers may be useful for predicting DME in older T2DM patients.
Collapse
Affiliation(s)
- Sang Youl Rhee
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Eun Sung Jung
- Department of Systems Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Dong Ho Suh
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Su Jin Jeong
- Statistics Support Department, Kyung Hee University Medical Center Medical Science Research Institute, Seoul, Republic of Korea
| | - Kiyoung Kim
- Department of Ophthalmology, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Suk Chon
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Seung-Young Yu
- Department of Ophthalmology, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Jeong-Taek Woo
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Choong Hwan Lee
- Department of Systems Biotechnology, Konkuk University, Seoul, Republic of Korea.
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
- Research Institute for Bioactive-Metabolome Network, Konkuk University, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Peng L, Sun B, Liu Y, Huang J, Chen G, Zhang X, Chen C, Wang D, Wang G. Increased lipoxygenase and decreased cytochrome P450s metabolites correlated with the incidence of diabetic nephropathy: Potential role of eicosanoids from metabolomics in type 2 diabetic patients. Clin Exp Pharmacol Physiol 2021; 48:679-685. [PMID: 33605471 DOI: 10.1111/1440-1681.13471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/21/2020] [Accepted: 01/12/2021] [Indexed: 12/22/2022]
Abstract
Diabetic nephropathy (DN) is the major cause of chronic kidney disease and end-stage renal disease. Previous studies have demonstrated that long-chain omega-3 polyunsaturated fatty acids (PUFAs) might have therapeutic potential in reducing proteinuria in DN. However, the local level of eicosanoids derived from PUFAs in the plasma of DN patients remains unclear. This work aims to study the eicosanoid profile difference in plasma of DN patients and type 2 diabetes (T2D) without DN. A total of 27 T2D patients with similar diabetic duration were recruited and divided into T2D+DN group and T2D+NDN (non-DN) group based on urinary albumin excretion (UAE) detection. Using LC-MS/MS-based metabolomics, DN patients showed increased level of lipoxygenase (LOX) metabolites (5-HETE and LTB4) and decreased levels of eicosanoids derived according to the cytochrome P450s (CYP450) metabolic pathway (5,6-DHET; 14,15-DHET and 9,10-diHOME). Receiver operating characteristics and logistic regression analysis revealed increased level LOX metabolites and decreased level of CYP450 metabolites were significantly correlated with the incidence of DN in T2D patients. LOX and CYP450 metabolites correlated with DN incidence in T2D patients, which might be treatment targets for DN in T2D patients.
Collapse
Affiliation(s)
- Liyuan Peng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Yajin Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Jing Huang
- Tianjin Key Laboratory of Metabolic Diseases, Key Laboratory of Immune Microenvironment and Disease-Ministry of Education, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Guangzhi Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Key Laboratory of Immune Microenvironment and Disease-Ministry of Education, Department of Physiology and Pathophysiology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Daowen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
34
|
Eicosanoid blood vessel regulation in physiological and pathological states. Clin Sci (Lond) 2021; 134:2707-2727. [PMID: 33095237 DOI: 10.1042/cs20191209] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/26/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022]
Abstract
Arachidonic acid can be metabolized in blood vessels by three primary enzymatic pathways; cyclooxygenase (COX), lipoxygenase (LO), and cytochrome P450 (CYP). These eicosanoid metabolites can influence endothelial and vascular smooth muscle cell function. COX metabolites can cause endothelium-dependent dilation or constriction. Prostaglandin I2 (PGI2) and thromboxane (TXA2) act on their respective receptors exerting opposing actions with regard to vascular tone and platelet aggregation. LO metabolites also influence vascular tone. The 12-LO metabolite 12S-hydroxyeicosatrienoic acid (12S-HETE) is a vasoconstrictor whereas the 15-LO metabolite 11,12,15-trihydroxyeicosatrienoic acid (11,12,15-THETA) is an endothelial-dependent hyperpolarizing factor (EDHF). CYP enzymes produce two types of eicosanoid products: EDHF vasodilator epoxyeicosatrienoic acids (EETs) and the vasoconstrictor 20-HETE. The less-studied cross-metabolites generated from arachidonic acid metabolism by multiple pathways can also impact vascular function. Likewise, COX, LO, and CYP vascular eicosanoids interact with paracrine and hormonal factors such as the renin-angiotensin system and endothelin-1 (ET-1) to maintain vascular homeostasis. Imbalances in endothelial and vascular smooth muscle cell COX, LO, and CYP metabolites in metabolic and cardiovascular diseases result in vascular dysfunction. Restoring the vascular balance of eicosanoids by genetic or pharmacological means can improve vascular function in metabolic and cardiovascular diseases. Nevertheless, future research is necessary to achieve a more complete understanding of how COX, LO, CYP, and cross-metabolites regulate vascular function in physiological and pathological states.
Collapse
|
35
|
Wang B, Wu L, Chen J, Dong L, Chen C, Wen Z, Hu J, Fleming I, Wang DW. Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets. Signal Transduct Target Ther 2021; 6:94. [PMID: 33637672 PMCID: PMC7910446 DOI: 10.1038/s41392-020-00443-w] [Citation(s) in RCA: 592] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/04/2020] [Accepted: 10/15/2020] [Indexed: 01/31/2023] Open
Abstract
The arachidonic acid (AA) pathway plays a key role in cardiovascular biology, carcinogenesis, and many inflammatory diseases, such as asthma, arthritis, etc. Esterified AA on the inner surface of the cell membrane is hydrolyzed to its free form by phospholipase A2 (PLA2), which is in turn further metabolized by cyclooxygenases (COXs) and lipoxygenases (LOXs) and cytochrome P450 (CYP) enzymes to a spectrum of bioactive mediators that includes prostanoids, leukotrienes (LTs), epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid (diHETEs), eicosatetraenoic acids (ETEs), and lipoxins (LXs). Many of the latter mediators are considered to be novel preventive and therapeutic targets for cardiovascular diseases (CVD), cancers, and inflammatory diseases. This review sets out to summarize the physiological and pathophysiological importance of the AA metabolizing pathways and outline the molecular mechanisms underlying the actions of AA related to its three main metabolic pathways in CVD and cancer progression will provide valuable insight for developing new therapeutic drugs for CVD and anti-cancer agents such as inhibitors of EETs or 2J2. Thus, we herein present a synopsis of AA metabolism in human health, cardiovascular and cancer biology, and the signaling pathways involved in these processes. To explore the role of the AA metabolism and potential therapies, we also introduce the current newly clinical studies targeting AA metabolisms in the different disease conditions.
Collapse
Affiliation(s)
- Bei Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Lujin Wu
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Jing Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China
| | - Jiong Hu
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Hubei Province, Wuhan, China.
| |
Collapse
|
36
|
Otto M, Bucher C, Liu W, Müller M, Schmidt T, Kardell M, Driessen MN, Rossaint J, Gross ER, Wagner NM. 12(S)-HETE mediates diabetes-induced endothelial dysfunction by activating intracellular endothelial cell TRPV1. J Clin Invest 2021; 130:4999-5010. [PMID: 32584793 DOI: 10.1172/jci136621] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Patients with diabetes develop endothelial dysfunction shortly after diabetes onset that progresses to vascular disease underlying the majority of diabetes-associated comorbidities. Increased lipid peroxidation, mitochondrial calcium overload, and mitochondrial dysfunction are characteristics of dysfunctional endothelial cells in diabetic patients. We here identified that targeting the lipid peroxidation product 12(S)-hydroxyeicosatetraenoic acid-induced [12(S)-HETE-induced] activation of the intracellularly located cation channel transient receptor potential vanilloid 1 (TRPV1) in endothelial cells is a means to causally control early-stage vascular disease in type I diabetic mice. Mice with an inducible, endothelium-specific 12/15-lipoxygenase (12/15Lo) knockout were protected similarly to TRPV1-knockout mice from type 1 diabetes-induced endothelial dysfunction and impaired vascular regeneration following arterial injury. Both 12(S)-HETE in concentrations found in diabetic patients and TRPV1 agonists triggered mitochondrial calcium influx and mitochondrial dysfunction in endothelial cells, and 12(S)-HETE effects were absent in endothelial cells from TRPV1-knockout mice. As a therapeutic consequence, we found that a peptide targeting 12(S)-HETE-induced TRPV1 interaction at the TRPV1 TRP box ameliorated diabetes-induced endothelial dysfunction and augmented vascular regeneration in diabetic mice. Our findings suggest that pharmacological targeting of increased endothelial lipid peroxidation can attenuate diabetes-induced comorbidities related to vascular disease.
Collapse
Affiliation(s)
- Mandy Otto
- Department of Anesthesiology, Intensive Care and Pain Medicine, and
| | - Clarissa Bucher
- Department of Anesthesiology, Intensive Care and Pain Medicine, and
| | - Wantao Liu
- Department of Anesthesiology, Intensive Care and Pain Medicine, and
| | - Melanie Müller
- Department of Anesthesiology, Intensive Care and Pain Medicine, and
| | - Tobias Schmidt
- Department of Anesthesiology, Intensive Care and Pain Medicine, and.,Institute of Physiology I, University Hospital Münster, Münster, Germany
| | - Marina Kardell
- Department of Anesthesiology, Intensive Care and Pain Medicine, and
| | | | - Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, and
| | - Eric R Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, California, USA
| | | |
Collapse
|
37
|
Muzaffar S, Shahid W, Riaz N, Saleem M, Ashraf M, Aziz-Ur-Rehman, Bashir B, Kaleem A, Al-Rashida M, Baral B, Bhattarai K, Gross H. Probing phenylcarbamoylazinane-1,2,4-triazole amides derivatives as lipoxygenase inhibitors along with cytotoxic, ADME and molecular docking studies. Bioorg Chem 2021; 107:104525. [PMID: 33317840 DOI: 10.1016/j.bioorg.2020.104525] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/24/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022]
Abstract
Hunting small molecules as anti-inflammatory agents/drugs is an expanding and successful approach to treat several inflammatory diseases such as cancer, asthma, arthritis, and psoriasis. Besides other methods, inflammatory diseases can be treated by lipoxygenase inhibitors, which have a profound influence on the development and progression of inflammation. In the present study, a series of new N-alkyl/aralky/aryl derivatives (7a-o) of 2-(4-phenyl-5-(1-phenylcarbamoyl)piperidine-4H-1,2,4-triazol-3-ylthio)acetamide was synthesized and screened for their inhibitory potential against the enzyme 15-lipoxygenase. The simple precursor ethyl piperidine-4-carboxylate (a) was successively converted into phenylcarbamoyl derivative (1), hydrazide (2), semicarbazide (3) and N-phenylated 5-(1-phenylcarbamoyl)piperidine-1,2,4-triazole (4), then in combination with electrophiles (6a-o) through further multistep synthesis, final products (7a-o) were generated. All the synthesized compounds were characterized by FTIR, 1H, 13C NMR spectroscopy, EIMS, and HREIMS spectrometry. Almost all the synthesized compounds showed excellent inhibitory potential against the tested enzyme. Compounds 7c, 7f, 7d, and 7g displayed potent inhibitory potential (IC50 9.25 ± 0.26 to 21.82 ± 0.35 µM), followed by the compounds 7n, 7h, 7e, 7a, 7b, 7l, and 7o with IC50 values in the range of 24.56 ± 0.45 to 46.91 ± 0.57 µM. Compounds 7c, 7f, 7d exhibited 71.5 to 83.5% cellular viability by MTT assay compared with standard curcumin (76.9%) when assayed at 0.125 mM concentration. In silico ADME studies supported the drug-likeness of most of the molecules. In vitro inhibition studies were substantiated by molecular docking wherein the phenyl group attached to the triazole ring was making a π-δ interaction with Leu607. This work reveals the possibility of a synthetic approach of compounds in relation to lipoxygenase inhibition as potential lead compounds in drug discovery.
Collapse
Affiliation(s)
- Saima Muzaffar
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Wardah Shahid
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Naheed Riaz
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.
| | - Muhammad Saleem
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Muhammad Ashraf
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.
| | - Aziz-Ur-Rehman
- Department of Chemistry, Government College University Lahore, Lahore 54000, Pakistan
| | - Bushra Bashir
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Ayesha Kaleem
- Department of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Mariya Al-Rashida
- Department of Chemistry, Forman Christian College (A Chartered University), Ferozepur Road Lahore, Lahore 54600, Pakistan
| | - Bikash Baral
- Department of Biochemistry, University of Turku, Tykistökatu 6, Finland
| | - Keshab Bhattarai
- Department of Pharmaceutical Biology, Auf der Morgenstelle 8, 72076, University of Tuebingen, Tuebingen, Germany
| | - Harald Gross
- Department of Pharmaceutical Biology, Auf der Morgenstelle 8, 72076, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
38
|
Xue Y, Deng Q, Zhang Q, Ma Z, Chen B, Yu X, Peng H, Yao S, Liu J, Ye Y, Pan G. Gigantol ameliorates CCl 4-induced liver injury via preventing activation of JNK/cPLA2/12-LOX inflammatory pathway. Sci Rep 2020; 10:22265. [PMID: 33335297 PMCID: PMC7746690 DOI: 10.1038/s41598-020-79400-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
Arachidonic acid (AA) signaling pathway is an important constituent of inflammatory processes. In our previous study, it was found that dihydro-stilbene gigantol relieved hepatic inflammation in mice with CCl4-induced acute liver injury. This study aimed to investigate the involvement of arachidonate metabolic cascade in this process. Our results showed CCl4 activated AA metabolism with the evidence of cPLA2 phosphorylation, which was dependent on the MAPK/JNK activation. Pretreatment with JNK inhibitor SU3327 or gigantol abolished the cPLA2 activation, along with the attenuation of liver damage. Besides, gigantol markedly decreased immune cells activation. Metabolomic analysis revealed that gigantol universally reversed the upregulation of major AA metabolites in injured mouse livers induced by CCl4, especially 12-hydroxyeicosatetraenoic acid (12-HETE). Gigantol also decreased the mRNA and protein expression of platelet-, and leukocyte-type 12-lipoxxygenase (LOX) in the liver. Furthermore, pan-LOX inhibitor nordihydroguaiaretic acid (NDGA) and specific 12-LOX inhibitors baicalein and ML351 attenuated the liver injury to the same extent as gigantol. Overall, our study elucidated a comprehensive profile of AA metabolites during hepatic inflammation caused by CCl4, highlighting the role of 12-LOX-12-HETE pathway in this process. And gigantol alleviated liver inflammation partly through inhibiting the JNK/cPLA2/12-LOX pathway.
Collapse
Affiliation(s)
- Yaru Xue
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiangqiang Deng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qingli Zhang
- Institutional Technology Service Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhenghua Ma
- State Key Laboratory of Drug Research and Natural Products Chemistry Department Shanghai, Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,SIMM-CUHK Joint Research Laboratory for Promoting Globalization of Traditional Chinese Medicines, Shanghai, 201203, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, 201203, China
| | - Binfan Chen
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaolu Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huige Peng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Sheng Yao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,State Key Laboratory of Drug Research and Natural Products Chemistry Department Shanghai, Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.,SIMM-CUHK Joint Research Laboratory for Promoting Globalization of Traditional Chinese Medicines, Shanghai, 201203, China
| | - Jia Liu
- Institutional Technology Service Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yang Ye
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China. .,State Key Laboratory of Drug Research and Natural Products Chemistry Department Shanghai, Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China. .,SIMM-CUHK Joint Research Laboratory for Promoting Globalization of Traditional Chinese Medicines, Shanghai, 201203, China. .,School of Life Science and Technology, Shanghai Tech University, Shanghai, 201203, China.
| | - Guoyu Pan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
39
|
Ali M, Bakr MH, Abdelzaher LA, Sayed SA, Mali V, Desai AA, Radwan E. Lisofylline mitigates cardiac inflammation in a mouse model of obesity through improving insulin secretion and activating cardiac AMPK signaling pathway. Cytokine 2020; 138:155398. [PMID: 33341003 DOI: 10.1016/j.cyto.2020.155398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 12/20/2022]
Abstract
Obesity has emerged as a leading cause of death in the last few decades, mainly due to associated cardiovascular diseases. Obesity, inflammation, and insulin resistance are strongly interlinked. Lisofylline (LSF), an anti-inflammatory agent, demonstrated protection against type 1 diabetes, as well as reduced obesity-induced insulin resistance and adipose tissue inflammation. However, its role in mitigating cardiac inflammation associated with obesity is not well studied. Mice were divided into 4 groups; the first group was fed regular chow diet, the second was fed regular chow diet and treated with LSF, the third was fed high fat diet (HFD), and the fourth was fed HFD and treated with LSF. Cardiac inflammation was interrogated via expression levels of TNF α, interleukins 6 and 10, phosphorylated STAT4 and lipoxygenases 12 and 12/15. Apoptosis and expression of the survival gene, AMPK, were also evaluated. We observed that LSF alleviated obesity-induced cardiac injury indirectly by improving both pancreatic β-cell function and insulin sensitivity, as well as, directly via upregulation of cardiac AMPK expression and downregulation of cardiac inflammation and apoptosis. LSF may represent an effective therapy targeting obesity-induced metabolic and cardiovascular complications.
Collapse
Affiliation(s)
- Maha Ali
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Marwa H Bakr
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Lobna A Abdelzaher
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Sally A Sayed
- Department of Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Vishal Mali
- Krannert Institute of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Ankit A Desai
- Krannert Institute of Cardiology, Department of Medicine, Indiana University, Indianapolis, IN, United States
| | - Eman Radwan
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
40
|
Leuti A, Fazio D, Fava M, Piccoli A, Oddi S, Maccarrone M. Bioactive lipids, inflammation and chronic diseases. Adv Drug Deliv Rev 2020; 159:133-169. [PMID: 32628989 DOI: 10.1016/j.addr.2020.06.028] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/09/2020] [Accepted: 06/25/2020] [Indexed: 02/08/2023]
Abstract
Endogenous bioactive lipids are part of a complex network that modulates a plethora of cellular and molecular processes involved in health and disease, of which inflammation represents one of the most prominent examples. Inflammation serves as a well-conserved defence mechanism, triggered in the event of chemical, mechanical or microbial damage, that is meant to eradicate the source of damage and restore tissue function. However, excessive inflammatory signals, or impairment of pro-resolving/anti-inflammatory pathways leads to chronic inflammation, which is a hallmark of chronic pathologies. All main classes of endogenous bioactive lipids - namely eicosanoids, specialized pro-resolving lipid mediators, lysoglycerophopsholipids and endocannabinoids - have been consistently involved in the chronic inflammation that characterises pathologies such as cancer, diabetes, atherosclerosis, asthma, as well as autoimmune and neurodegenerative disorders and inflammatory bowel diseases. This review gathers the current knowledge concerning the involvement of endogenous bioactive lipids in the pathogenic processes of chronic inflammatory pathologies.
Collapse
|
41
|
Dong C, Liu S, Cui Y, Guo Q. 12-Lipoxygenase as a key pharmacological target in the pathogenesis of diabetic nephropathy. Eur J Pharmacol 2020; 879:173122. [DOI: 10.1016/j.ejphar.2020.173122] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/07/2020] [Accepted: 04/17/2020] [Indexed: 12/18/2022]
|
42
|
Chen Y, Zhou B, Yu Z, Yuan P, Sun T, Gong J, Zhang Y, Wang T, Wang S, Liu K, Liu J. Baicalein Alleviates Erectile Dysfunction Associated With Streptozotocin-Induced Type I Diabetes by Ameliorating Endothelial Nitric Oxide Synthase Dysfunction, Inhibiting Oxidative Stress and Fibrosis. J Sex Med 2020; 17:1434-1447. [PMID: 32586748 DOI: 10.1016/j.jsxm.2020.04.390] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Management of diabetes mellitus induced-erectile dysfunction (DMED) is challenging because of its poor responses to phosphodiesterase type 5 inhibitors. Increasingly important roles of 12-lipoxygenase (12-LOX) have been proven in diabetes mellitus. AIM To investigate 12-LOX activity and therapeutic effect of its inhibitor, baicalein (BE), on DMED. METHODS Intraperitoneal streptozotocin injection was used to induce type I DM, and an apomorphine test was used to evaluate erectile function. In experiment A, we assessed 12-LOX expression alteration in the corpus cavernosum (CC) of rats with DMED of different levels of severity. In experiment B, rats with DMED were intraperitoneally injected with BE for 4 weeks, and control rats were injected with vehicles. The erectile function was tested by cavernous nerve stimulation before penile tissue was harvested. We performed Western blot, immunohistochemistry, immunofluorescence, Masson trichrome staining, and enzyme-linked immunosorbent assays to measure related proteins in CC. MAIN OUTCOME MEASURE The main outcome measures included rectile response, histologic examination, and expression alteration of related proteins. RESULTS 12-LOX upregulation was associated with the progression of type I DMED. After 4 weeks treatment, compared with the DMED group, the DMED + BE group showed better erectile responses to cavernous nerve stimulation. In the DMED + BE group, significantly enhanced endothelial nitric oxide synthase/nitric oxide/cyclic guanosine monophosphate pathway, reduced 12-LOX expression, and inhibited p38 mitogen-activated protein kinase/arginase II/L-arginine pathway were showed in CC relative to the DMED group. In addition, overactivated oxidative stress and fibrosis in the DMED group were both partially ameliorated in the DMED + BE group. CLINICAL IMPLICATIONS BE may be considered as an effective therapy for DMED, but needs to be verified in future human investigations. STRENGTHS & LIMITATIONS The role of 12-LOX and its inhibitor, BE, is firstly demonstrated in rats with type I DMED. However, the experimental data are derived from animal models with without evidences from cellular-based experiments. CONCLUSION 12-LOX might serve as an important factor in the pathogenesis of type I DMED. BE alleviated erectile dysfunction in rats with type I DMED probably by inhibiting 12-LOX expression, ameliorating endothelial nitric oxide synthase dysfunction, as well as suppressing oxidative stress and fibrosis. Chen Y, Zhou B, Yu Z, et al. Baicalein Alleviates Erectile Dysfunction Associated With Streptozotocin-Induced Type I Diabetes by Ameliorating Endothelial Nitric Oxide Synthase Dysfunction, Inhibiting Oxidative Stress and Fibrosis. J Sex Med 2020;17:1434-1447.
Collapse
Affiliation(s)
- Yinwei Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bingyan Zhou
- Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhe Yu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Penghui Yuan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Taotao Sun
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianan Gong
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shaogang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kang Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - JiHong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
43
|
Díaz-Gerevini GT, Daín A, Pasqualini ME, López CB, Eynard AR, Repossi G. Diabetic encephalopathy: beneficial effects of supplementation with fatty acids ω3 and nordihydroguaiaretic acid in a spontaneous diabetes rat model. Lipids Health Dis 2019; 18:43. [PMID: 30736810 PMCID: PMC6368734 DOI: 10.1186/s12944-018-0938-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 11/30/2018] [Indexed: 02/07/2023] Open
Abstract
Background Diabetic encephalopathy is a chronic complications of diabetes mellitus that affects the central nervous system. We evaluated the effect of ω3 and ω6 polyunsaturated fatty acids (PUFAs) supplementation plus the antioxidant agent nordihydroguaiaretic acid (NDGA) on the etiopathology of diabetic encephalopathy in eSS rats, a spontaneous model of type 2 diabetes. Methods One hundred twenty spontaneous diabetic eSS male rats and 38 non-diabetic Wistar, used as healthy control, received monthly by intraperitoneal route, ω3 or ω6 PUFA (6.25 mg/kg) alone or plus NDGA (1.19 mg/kg) for 12 months. Diabetic rats had a worse performance in behavioural Hole-Board test. Histopathological analysis confirmed lesions in diabetic rats brain tissues. We also detected low expression of synaptophysin, a protein linked to release of neurotransmitters, by immunohistochemically techniques in eSS rats brain. Biochemical and histopathological studies of brain were performed at 12th month. Biochemical analysis showed altered parameters related to metabolism. High levels of markers of oxidative stress and inflammation were detected in plasma and brain tissues. Data were analysed by ANOVA test and paired t test was used by comparison of measurements of the same parameter at different times. Results The data obtained in this work showed that behavioural, biochemical and morphological alterations observed in eSS rats are compatible with previously reported indices in diabetic encephalopathy and are associated with increased glucolipotoxicity, chronic low-grade inflammation and oxidative stress burden. Experimental treatments assayed modulated the values of studied parameters. Conclusions The treatments tested with ω3 or ω3 plus NDGA showed improvement in the values of the studied parameters in eSS diabetic rats. These observations may form the basis to help in prevent and manage the diabetic encephalopathy. Electronic supplementary material The online version of this article (10.1186/s12944-018-0938-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gustavo Tomás Díaz-Gerevini
- Biología Celular, Histología y Embriología. Facultad de Ciencias Médicas, INICSA CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.,Geriatric Center "San Ricardo Pampuri", Villa Carlos Paz and Gerontology Committee, Argentine Society of Diabetes, Córdoba, Argentina
| | - Alejandro Daín
- Biología Celular, Histología y Embriología. Facultad de Ciencias Médicas, INICSA CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Eugenia Pasqualini
- Biología Celular, Histología y Embriología. Facultad de Ciencias Médicas, INICSA CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Cristina B López
- Biología Celular, Histología y Embriología. Facultad de Ciencias Médicas, INICSA CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.,Cátedra de Histología y Embriología, Universidad Nacional de La Rioja (UNLaR), La Rioja, Argentina
| | - Aldo R Eynard
- Biología Celular, Histología y Embriología. Facultad de Ciencias Médicas, INICSA CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gastón Repossi
- Biología Celular, Histología y Embriología. Facultad de Ciencias Médicas, INICSA CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|