1
|
Liu M, Du X, Chen H, Bai C, Lan L. Systemic investigation of di-isobutyl phthalate (DIBP) exposure in the risk of cardiovascular via influencing the gut microbiota arachidonic acid metabolism in obese mice model. Regen Ther 2024; 27:290-300. [PMID: 38638558 PMCID: PMC11024931 DOI: 10.1016/j.reth.2024.03.024] [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: 02/02/2024] [Revised: 03/17/2024] [Accepted: 03/24/2024] [Indexed: 04/20/2024] Open
Abstract
Phthalate esters (PE), a significant class of organic compounds used in industry, can contaminate humans and animals by entering water and food chains. Recent studies demonstrate the influence of PE on the development and progression of heart diseases, particularly in obese people. Di-isobutyl phthalate (DIBP) was administered orally to normal and diet-induced obese mice in this research to assess cardiovascular risk. The modifications in the microbial composition and metabolites were examined using RNA sequencing and mass spectrometry analysis. Based on the findings, lean group rodents were less susceptible to DIBP exposure than fat mice because of their cardiovascular systems. Histopathology examinations of mice fed a high-fat diet revealed lesions and plagues that suggested a cardiovascular risk. In the chronic DIBP microbial remodeling metagenomics Faecalibaculum rodentium was the predominant genera in obese mice. According to metabolomics data, arachidonic acid (AA) metabolism changes caused by DIBP were linked to unfavorable cardiovascular events. Our research offers new understandings of the cardiovascular damage caused by DIBP exposure in obese people and raises the possibility that arachidonic acid metabolism could be used as a regulator of the gut microbiota to avert related diseases.
Collapse
Affiliation(s)
- Min Liu
- Department of General Practice, First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Taiyuan, Shanxi, 030001, China
| | - Xifeng Du
- Department of General Practice, First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Taiyuan, Shanxi, 030001, China
| | - Huifang Chen
- Department of General Practice, First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Taiyuan, Shanxi, 030001, China
| | - Chenkai Bai
- Department of General Practice, First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Taiyuan, Shanxi, 030001, China
| | - Lizhen Lan
- Department of General Practice, First Hospital of Shanxi Medical University, No.85, Jiefang South Road, Taiyuan, Shanxi, 030001, China
| |
Collapse
|
2
|
Men JL, Xue YJ, Fu Y, Bai X, Wang XB, Zhou HL. Decoding the role of HIF-1α in immunoregulation in Litopenaeus vannamei under hypoxic stress. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109962. [PMID: 39396558 DOI: 10.1016/j.fsi.2024.109962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 09/10/2024] [Accepted: 10/09/2024] [Indexed: 10/15/2024]
Abstract
Hypoxia poses a significant challenge to aquatic organisms, especially Litopenaeus vannamei (L. vannamei), which play a vital role in the global aquaculture industry. Hypoxia-inducible factor 1α (HIF-1α) is a pivotal regulator of the organism's adaptation to hypoxic conditions. To understand of how HIF-1α affects the immunity of L. vannamei under hypoxic conditions, we conducted a thorough study involving various approaches. These included observing tissue morphology, analyzing the expression of immune-related genes, assessing the activities of immune-related enzymes, and exploring immune-related pathways. Our study revealed that RNA interference (RNAi)-mediated knockdown of HIF-1α markedly reduced HIF-1α expression in the gill (75-95 %), whereas the reduction ranged from 2 to 43 % in the hepatopancreas. Knockdown of HIF-1α resulted in increased damage to both gill and hepatopancreatic tissues in hypoxic conditions. Additionally, immune-related genes, including Astakine (AST), Hemocyanin (HC), and Ferritin (FT), as well as immune-related enzymes such as Acid Phosphatase (ACP), Alkaline Phosphatase (AKP), and Phenoloxidase (PO), exhibited intricate regulatory patterns in response to hypoxia stress following the knockdown of HIF-1α. Transcriptome analysis revealed that HIF-1α knockdown significantly impacts multiple signaling pathways, including the JAK-STAT signaling pathway, Th17 cell differentiation pathways, PI3K-Akt signaling pathway, ErbB signaling pathway, MAPK signaling pathway, chemokine signaling pathway, ribosomal pathways, apoptosis, lysosomes and arachidonic acid metabolism. These alterations disrupt the organism's immune balance and interfere with normal metabolic processes, potentially leading to various immune-related diseases. We speculate that the weakened immune response resulting from HIF-1 inhibition is due to the reduced metabolic capacity, and the existence of a direct regulatory relationship between them requires further exploration. This study greatly advances our understanding of the vital role that HIF-1α plays in regulating immune responses in shrimp under hypoxic conditions, thereby deepening our comprehension of this critical biological mechanism.
Collapse
Affiliation(s)
- Jia L Men
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Yi J Xue
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Ying Fu
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Xue Bai
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Xiao B Wang
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Hai L Zhou
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China; One Health Institute, Hainan University, Haikou, Hainan, 570228, China; State Key Laboratory of South China Sea Marine Resource Utilization, Hainan University, Haikou, 570228, China.
| |
Collapse
|
3
|
Kou Y, Zhang S, Chen J, Shen Y, Zhang Z, Huang H, Ma Y, Xiang Y, Liao L, Zhou J, Cheng W, Zhou Y, Yang H, Liu Z, Wei Y, Wang H, Wang Y. A mouse protozoan boosts antigen-specific mucosal IgA responses in a specific lipid metabolism- and signaling-dependent manner. Nat Commun 2024; 15:7914. [PMID: 39256385 PMCID: PMC11387640 DOI: 10.1038/s41467-024-52336-z] [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: 07/06/2023] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
IgA antibodies play an important role in mucosal immunity. However, there is still no effective way to consistently boost mucosal IgA responses, and the factors influencing these responses are not fully understood. We observed that colonization with the murine intestinal symbiotic protozoan Tritrichomonas musculis (T.mu) boosted antigen-specific mucosal IgA responses in wild-type C57BL/6 mice. This enhancement was attributed to the accumulation of free arachidonic acid (ARA) in the intestinal lumen, which served as a signal to stimulate the production of antigen-specific mucosal IgA. When ARA was prevented from undergoing its downstream metabolic transformation using the 5-lipoxygenase inhibitor zileuton or by blocking its downstream biological signaling through genetic deletion of the Leukotriene B4 receptor 1 (Blt1), the T.mu-mediated enhancement of antigen-specific mucosal IgA production was suppressed. Moreover, both T.mu transfer and dietary supplementation of ARA augmented the efficacy of an oral vaccine against Salmonella infection, with this effect being dependent on Blt1. Our findings elucidate a tripartite circuit linking nutrients from the diet or intestinal microbiota, host lipid metabolism, and the mucosal humoral immune response.
Collapse
Affiliation(s)
- Yanbo Kou
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Shenghan Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
- Department of Central Laboratory, Xuzhou Central Hospital, Xuzhou, China
| | - Junru Chen
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yusi Shen
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Zhiwei Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Haohan Huang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yulu Ma
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yaoyao Xiang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Longxiang Liao
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Junyang Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Wanpeng Cheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yuan Zhou
- Xuzhou Key Laboratory of Laboratory Diagnostics, Medical Technology School, Xuzhou Medical University, Xuzhou, China
| | - Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, Medical Technology School, Xuzhou Medical University, Xuzhou, China
| | - Zhuanzhuan Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yanxia Wei
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Hui Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yugang Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China.
- Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
4
|
Dong Z, Wang Q. The causal relationship between human blood metabolites and risk of peripheral artery disease: a Mendelian randomization study. Front Cardiovasc Med 2024; 11:1435106. [PMID: 39318836 PMCID: PMC11420124 DOI: 10.3389/fcvm.2024.1435106] [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: 05/19/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
Background Peripheral Artery Disease (PAD) is a common vascular disorder typically caused by atherosclerosis, leading to impaired blood supply to the lower extremities, resulting in pain, necrosis, and even amputation. Despite extensive research into the pathogenesis of PAD, many mysteries remain, particularly regarding its association with human blood metabolites. Methods To explore the causal relationship between 1,400 serum metabolites and PAD, a two-sample Mendelian randomization (MR) analysis was conducted. The Inverse Variance-Weighted (IVW) method was the primary technique used to estimate the causal impact of the metabolites on PAD. To enhance the analysis, several additional methods were employed: MR-Egger regression, weighted median, simple mode, and weighted mode. These methods provided a comprehensive evaluation beyond the primary IVW estimation. To ensure the validity of the MR findings, sensitivity analysis was performed. Furthermore, a bidirectional MR approach was applied to explore the possibility of a reverse causal effect between PAD and potential candidate metabolites. Results After rigorous selection, significant associations were found between 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4) and X-17653 levels with PAD. 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4) was positively associated with increased PAD risk (IVW OR = 1.13, 95% CI, 1.06-1.21; P < 0.001). X-17653 levels were associated with decreased PAD risk (IVW OR = 0.88, 95% CI, 0.83-0.94; P < 0.001). In the reverse direction, PAD was positively associated with increased 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4) levels (IVW OR = 1.16, 95% CI, 1.01-1.34; P = 0.036). PAD was not associated with X-17653. Conclusion Among 1,400 blood metabolites, 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4) and X-17653 are significantly associated with PAD risk. Importantly, in the reverse direction, PAD was found to be positively associated with increased levels of 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4). This highlights the bidirectional nature of the association and suggests a potential feedback mechanism between PAD and this specific lipid species. 1-(1-enyl-stearoyl)-2-arachidonoyl-GPE (p-18:0/20:4) may serve as potential biomarkers for PAD, aiding early diagnosis and providing novel avenues for personalized treatment and management. However, further validation and research are warranted despite the promising results.
Collapse
Affiliation(s)
| | - Qingyun Wang
- Department of Cardiothoracic Surgery, Beijing Shunyi Hospital, Beijing, China
| |
Collapse
|
5
|
Stewart KS, Abdusselamoglu MD, Tierney MT, Gola A, Hur YH, Gonzales KAU, Yuan S, Bonny AR, Yang Y, Infarinato NR, Cowley CJ, Levorse JM, Pasolli HA, Ghosh S, Rothlin CV, Fuchs E. Stem cells tightly regulate dead cell clearance to maintain tissue fitness. Nature 2024; 633:407-416. [PMID: 39169186 PMCID: PMC11390485 DOI: 10.1038/s41586-024-07855-6] [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: 04/30/2023] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
Billions of cells are eliminated daily from our bodies1-4. Although macrophages and dendritic cells are dedicated to migrating and engulfing dying cells and debris, many epithelial and mesenchymal tissue cells can digest nearby apoptotic corpses1-4. How these non-motile, non-professional phagocytes sense and eliminate dying cells while maintaining their normal tissue functions is unclear. Here we explore the mechanisms that underlie their multifunctionality by exploiting the cyclical bouts of tissue regeneration and degeneration during hair cycling. We show that hair follicle stem cells transiently unleash phagocytosis at the correct time and place through local molecular triggers that depend on both lipids released by neighbouring apoptotic corpses and retinoids released by healthy counterparts. We trace the heart of this dual ligand requirement to RARγ-RXRα, whose activation enables tight regulation of apoptotic cell clearance genes and provides an effective, tunable mechanism to offset phagocytic duties against the primary stem cell function of preserving tissue integrity during homeostasis. Finally, we provide functional evidence that hair follicle stem cell-mediated phagocytosis is not simply redundant with professional phagocytes but rather has clear benefits to tissue fitness. Our findings have broad implications for other non-motile tissue stem or progenitor cells that encounter cell death in an immune-privileged niche.
Collapse
Affiliation(s)
- Katherine S Stewart
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA.
| | - Merve Deniz Abdusselamoglu
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Matthew T Tierney
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Anita Gola
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Yun Ha Hur
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Kevin A U Gonzales
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Department of Discovery Technology and Genomics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Shaopeng Yuan
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge, UK
| | - Alain R Bonny
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Yihao Yang
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Altos Labs, San Diego, CA, USA
| | - Nicole R Infarinato
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- PrecisionScienta, Yardley, PA, USA
| | - Christopher J Cowley
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John M Levorse
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Cardiovascular Research Group, Temple University, Philadelphia, PA, USA
| | - Hilda Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY, USA
| | - Sourav Ghosh
- Departments of Neurology and Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Carla V Rothlin
- Departments of Immunobiology and Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
6
|
Sun K, Meesapyodsuk D, Qiu X. Biosynthetic mechanisms of omega-3 polyunsaturated fatty acids in microalgae. J Biol Chem 2024; 300:107699. [PMID: 39173949 PMCID: PMC11418110 DOI: 10.1016/j.jbc.2024.107699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
Marine microalgae are the primary producers of ω3 polyunsaturated fatty acids (PUFAs), such as octadecapentaenoic acid (OPA, 18:5n-3) and docosahexaenoic acid (DHA, 22:6n-3) for food chains. However, the biosynthetic mechanisms of these PUFAs in the algae remain elusive. To study how these fatty acids are synthesized in microalgae, a series of radiolabeled precursors were used to trace the biosynthetic process of PUFAs in Emiliania huxleyi. Feeding the alga with 14C-labeled acetic acid in a time course showed that OPA was solely found in glycoglycerolipids such as monogalactosyldiacylglycerol (MGDG) and digalactosyldiacylglycerol (DGDG) synthesized plastidically by sequential desaturations while DHA was exclusively found in phospholipids synthesized extraplastidically. Feeding the alga with 14C-labeled α-linolenic acid (ALA), linoleic acid (LA), and oleic acid (OA) showed that DHA was synthesized extraplastidically from fed ALA and LA, but not from OA, implying that the aerobic pathway of DHA biosynthesis is incomplete with missing a Δ12 desaturation step. The in vitro enzymatic assays with 14C-labeled malonyl-CoA showed that DHA was synthesized from acetic acid by a PUFA synthase. These results provide the first and conclusive biochemistry evidence that OPA is synthesized by a plastidic aerobic pathway through sequential desaturations with the last step of Δ3 desaturation, while DHA is synthesized by an extraplastidic anaerobic pathway catalyzed by a PUFA synthase in the microalga.
Collapse
Affiliation(s)
- Kaiwen Sun
- Department of Food & Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | | - Xiao Qiu
- Department of Food & Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
7
|
Hanna V, Abd El-Ghany MN, Ibrahim MIM, Abdel-Rahman TM, Tallima H. Novel Approaches to Mortierella alpina Identification and Arachidonic Acid Production Optimization. ACS OMEGA 2024; 9:34456-34463. [PMID: 39157088 PMCID: PMC11325418 DOI: 10.1021/acsomega.4c02294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/16/2024] [Accepted: 06/19/2024] [Indexed: 08/20/2024]
Abstract
Arachidonic acid (ARA) is an integral constituent of cell structures and is instrumental for the nervous, muscular, and immune systems' functions. The sore need for this nutrient may be fulfilled via production based on the fungus Mortierella alpina. The identity of the M. alpina culture obtained from Assiut University, Egypt, was confirmed based on internal transcribed spacer DNA barcoding and elongation enzyme RNA sequencing. Liquid media glucose and peptone as carbon and nitrogen sources, respectively, and diverse micronutritional factors were adjusted for optimal biomass and ARA production. Shake flask cultivation at 25 °C for 7 days produced around 0.570 g of ARA per liter of culture. M. alpina treatment using mutagen 5-fluorouracil and octyl gallate-supplemented glucose-yeast-agar screening plates and shake-flask incubation at 25 °C, then at 20 °C, followed by aging at 10 °C, led to >3 g ARA/liter culture, a yield considered suitable for potential commercial production.
Collapse
Affiliation(s)
- Violette
S. Hanna
- Biotechnology
Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | | | - Mohamed I. M. Ibrahim
- Food
Toxicology and Contaminant Department, National
Research Centre, Giza 12622, Egypt
| | - Tahany M. Abdel-Rahman
- Botany
and Microbiology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Hatem Tallima
- Department
of Chemistry, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Cairo, Egypt
| |
Collapse
|
8
|
Lu W, Aihaiti A, Abudukeranmu P, Liu Y, Gao H. Arachidonic acid metabolism as a novel pathogenic factor in gastrointestinal cancers. Mol Cell Biochem 2024:10.1007/s11010-024-05057-2. [PMID: 38963615 DOI: 10.1007/s11010-024-05057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Gastrointestinal (GI) cancers are a major global health burden, representing 20% of all cancer diagnoses and 22.5% of global cancer-related deaths. Their aggressive nature and resistance to treatment pose a significant challenge, with late-stage survival rates below 15% at five years. Therefore, there is an urgent need to delve deeper into the mechanisms of gastrointestinal cancer progression and optimize treatment strategies. Increasing evidence highlights the active involvement of abnormal arachidonic acid (AA) metabolism in various cancers. AA is a fatty acid mainly metabolized into diverse bioactive compounds by three enzymes: cyclooxygenase, lipoxygenase, and cytochrome P450 enzymes. Abnormal AA metabolism and altered levels of its metabolites may play a pivotal role in the development of GI cancers. However, the underlying mechanisms remain unclear. This review highlights a unique perspective by focusing on the abnormal metabolism of AA and its involvement in GI cancers. We summarize the latest advancements in understanding AA metabolism in GI cancers, outlining changes in AA levels and their potential role in liver, colorectal, pancreatic, esophageal, gastric, and gallbladder cancers. Moreover, we also explore the potential of targeting abnormal AA metabolism for future therapies, considering the current need to explore AA metabolism in GI cancers and outlining promising avenues for further research. Ultimately, such investigations aim to improve treatment options for patients with GI cancers and pave the way for better cancer management in this area.
Collapse
Affiliation(s)
- Weiqin Lu
- General Surgery, Cancer Center, Department of Vascular Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | | | | | - Yajun Liu
- Aksu First People's Hospital, Xinjiang, China
| | - Huihui Gao
- Cancer Center, Department of Hospital Infection Management and Preventive Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Tallima H, Mahmoud SS. Mechanisms of Arachidonic Acid In Vitro Schistosomicidal Potential. ACS OMEGA 2024; 9:23316-23328. [PMID: 38854551 PMCID: PMC11154912 DOI: 10.1021/acsomega.3c09906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/23/2024] [Accepted: 03/28/2024] [Indexed: 06/11/2024]
Abstract
Arachidonic acid (ARA) was shown to possess safe and effective schistosomicidal impact on larval and adult Schistosoma mansoni and Schistosoma hematobium in vitro and in vivo in laboratory rodents and in children residing in low and high endemicity regions. We herein examine mechanisms underlying ARA schistosomicidal potential over two experiments, using in each pool a minimum of 50 adult male, female, or mixed-sex freshly recovered, ex vivo S. mansoni. Worms incubated in fetal calf serum-free medium were exposed to 0 or 10 mM ARA for 1 h at 37 °C and immediately processed for preparation of surface membrane and whole worm body homogenate extracts. Mixed-sex worms were additionally used for evaluating the impact of ARA exposure on the visualization of outer membrane cholesterol, sphingomyelin (SM), and ceramide in immunofluorescence assays. Following assessment of protein content, extracts of intact and ARA-treated worms were examined and compared for SM content, neutral sphingomyelinase activity, reactive oxygen species levels, and caspase 3/7 activity. Arachidonic acid principally led to perturbation of the organization, integrity, and SM content of the outer membrane of male and female worms and additionally impacted female parasites via stimulating neutral sphingomyelinase activity and oxidative stress. Arachidonic powerful action on female worms combined with its previously documented ovocidal activities supports its use as safe and effective therapy against schistosomiasis, provided implementation of the sorely needed and long waited-for chemical synthesis.
Collapse
Affiliation(s)
- Hatem Tallima
- Department
of Chemistry, School of Sciences and Engineering, American University in Cairo, New Cairo, Cairo 11835, Egypt
| | - Soheir S. Mahmoud
- Department
of Parasitology, Theodore Bilharz Research
Institute, Warrak El-Hadar, Imbaba,Giza 12411, Egypt
| |
Collapse
|
10
|
Roberts BM, Sczuroski CE, Caldwell AR, Zeppetelli DJ, Smith NI, Pecorelli VP, Gwin JA, Hughes JM, Staab JS. NSAIDs do not prevent exercise-induced performance deficits or alleviate muscle soreness: A placebo-controlled randomized, double-blinded, cross-over study. J Sci Med Sport 2024; 27:287-292. [PMID: 38383211 DOI: 10.1016/j.jsams.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/16/2024] [Accepted: 02/06/2024] [Indexed: 02/23/2024]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are frequently consumed by athletes to manage muscle soreness, expedite recovery, or improve performance. Despite the prevalence of NSAID use, their effects on muscle soreness and performance, particularly when administered prophylactically, remain unclear. This randomized, double-blind, counter-balanced, crossover study examined the effect of consuming a single dose of each of three NSAIDs (celecoxib, 200 mg; ibuprofen, 800 mg; flurbiprofen, 100 mg) or placebo 2 h before on muscle soreness and performance following an acute plyometric training session. Twelve healthy adults, aged 18-42 years, completed a standardized plyometric exercise session consisting of 10 sets of 10 repetitions at 40 % 1-repetition maximum (1RM) on a leg press device. During exercise, total work, rating of perceived exertion, and heart rate were measured. Maximum voluntary contraction force (MVC), vertical jump height, and muscle soreness were measured before exercise and 4-h and 24-h post-exercise. We found no significant differences in total work, heart rate, or rating of perceived exertion between treatments. Additionally, no significant differences in muscle soreness or vertical jump were observed between treatments. Ibuprofen and flurbiprofen did not prevent decrements in MVC, but celecoxib attenuated decreases in MVC 4-h post exercise (p < 0.05). This study suggests that athletes may not benefit from prophylactic ibuprofen or flurbiprofen treatment to prevent discomfort or performance decrements associated with exercise, but celecoxib may mitigate short-term performance decrements.
Collapse
Affiliation(s)
- Brandon M Roberts
- Military Performance Division, US Army Research Institute of Environmental Medicine, USA.
| | - Cara E Sczuroski
- Military Performance Division, US Army Research Institute of Environmental Medicine, USA
| | - Aaron R Caldwell
- Thermal and Mountain Medicine Division, US Army Research Institute of Environmental Medicine, USA
| | - David J Zeppetelli
- Military Performance Division, US Army Research Institute of Environmental Medicine, USA
| | - Nathaniel I Smith
- Military Performance Division, US Army Research Institute of Environmental Medicine, USA
| | - Vincent P Pecorelli
- Military Performance Division, US Army Research Institute of Environmental Medicine, USA
| | - Jess A Gwin
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, USA
| | - Julie M Hughes
- Military Performance Division, US Army Research Institute of Environmental Medicine, USA
| | - Jeffery S Staab
- Military Performance Division, US Army Research Institute of Environmental Medicine, USA
| |
Collapse
|
11
|
Li C, Xie R, Zhang S, Yun J, Zhong A, Cen Y, Chen J. Metabolism, fibrosis, and apoptosis: The effect of lipids and their derivatives on keloid formation. Int Wound J 2024; 21:e14733. [PMID: 38339798 PMCID: PMC10858330 DOI: 10.1111/iwj.14733] [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: 12/11/2023] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Keloids, pathological scars resulting from skin trauma, have traditionally posed significant clinical management challenges due to their persistence and high recurrence rates. Our research elucidates the pivotal roles of lipids and their derivatives in keloid development, driven by underlying mechanisms of abnormal cell proliferation, apoptosis, and extracellular matrix deposition. Key findings suggest that abnormalities in arachidonic acid (AA) synthesis and non-essential fatty acid synthesis are integral to keloid formation. Further, a complex interplay exists between lipid derivatives, notably butyric acid (BA), prostaglandin E2 (PGE2), prostaglandin D2 (PGD2), and the regulation of hyperfibrosis. Additionally, combinations of docosahexaenoic acid (DHA) with BA and 15-deoxy-Δ12,14-Prostaglandin J2 have exhibited pronounced cytotoxic effects. Among sphingolipids, ceramide (Cer) displayed limited pro-apoptotic effects in keloid fibroblasts (KFBs), whereas sphingosine 1-phosphate (S1P) was found to promote keloid hyperfibrosis, with its analogue, FTY720, demonstrating contrasting benefits. Both Vitamin D and hexadecylphosphorylcholine (HePC) showed potential antifibrotic and antiproliferative properties, suggesting their utility in keloid management. While keloids remain a prevalent concern in clinical practice, this study underscores the promising potential of targeting specific lipid molecules for the advancement of keloid therapeutic strategies.
Collapse
Affiliation(s)
- Chen‐yu Li
- Department of Burn and Plastic SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Ru‐xin Xie
- Department of Burn and Plastic SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Shi‐wei Zhang
- Department of Burn and Plastic SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Jiao Yun
- Department of Burn and Plastic SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Ai Zhong
- Department of Burn and Plastic SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Ying Cen
- Department of Burn and Plastic SurgeryWest China Hospital of Sichuan UniversityChengduChina
| | - Jun‐jie Chen
- Department of Burn and Plastic SurgeryWest China Hospital of Sichuan UniversityChengduChina
| |
Collapse
|
12
|
Qian HQ, Wu DC, Li CY, Liu XR, Han XK, Peng Y, Zhang H, Zhao BY, Zhao Y. A systematic review of traditional uses, phytochemistry, pharmacology and toxicity of Epimedium koreanum Nakai. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116957. [PMID: 37544344 DOI: 10.1016/j.jep.2023.116957] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/08/2023] [Accepted: 07/21/2023] [Indexed: 08/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Epimedium koreanum Nakai (E. koreanum), a member of the genus Epimedium in the family Berberidaceae, is a well-known and well-liked traditional herb used as a "kidney tonic". For thousands of years, it has been utilized for renal yang deficiency, impotence, spermatorrhea, impotence, weakness of tendons and bones, rheumatic paralysis and discomfort, numbness, and constriction. AIM OF THE STUDY The paper aims to comprehensively in-depth, and methodically review the most recent research on the traditional uses, phytochemistry, pharmacology, and toxicity of E. koreanum. MATERIALS AND METHODS Scientific databases including Web of Science, PubMed, Google Scholar, Elsevier, Springer, ScienceDirect, Baidu Scholar, and CNKI and medicine books in China were searched for relevant information on E. koreanum. RESULTS In traditional uses, E. koreanum is frequently used to treat various diseases like erectile dysfunction, infertility, rheumatoid arthritis, osteoporosis, asthma, kidney-yang deficiency syndrome, etc. To date, more than 379 compounds have been discovered from various parts of E. koreanum, including flavonoids, lignans, organic acids, terpenoids, hydrocarbons, dihydrophenanthrene derivatives, alkaloids, and others. Research has revealed that the compounds and crude extracts have a wide range of pharmacological effects on the reproductive, cardiovascular, and nervous systems, as well as anti-osteoporosis, anti-tumor, antioxidant, anti-inflammatory, immunomodulatory, hepatoprotective, and antiviral properties. Besides, the crude extracts show potential hepatotoxicity. CONCLUSION Based on recent domestic and international research investigations, E. koreanum contains a wealth of chemical components with pronounced pharmacological activities. Its traditional uses are numerous, and the majority of these traditional uses have been supported by contemporary pharmacological investigations. Crude extracts, on the other hand, can result in hepatotoxicity. Therefore, additional in vivo and in vitro experimental research on the pharmacology and toxicology of E. koreanum are required in the future to assess its safety and efficacy. This will give a firmer scientific foundation for its safe application and the development of new drugs in the future.
Collapse
Affiliation(s)
- Hui-Qin Qian
- Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China
| | - Dou-Can Wu
- Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China
| | - Chun-Yan Li
- Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China
| | - Xin-Ran Liu
- Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China
| | - Xin-Ke Han
- Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China
| | - Yuan Peng
- Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China
| | - Han Zhang
- Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China
| | - Bing-Yan Zhao
- Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China
| | - Yuan Zhao
- Sanquan College of Xinxiang Medical University, Xinxiang, 453000, China.
| |
Collapse
|
13
|
Bahadır Semerci A, Yıldırım M, Oztay F, Sagıroglu M, Tunç K. Evaluation of Fatty Acid Contents and Biological Activities of Jurinea turcica. Chem Biodivers 2024; 21:e202300084. [PMID: 38010957 DOI: 10.1002/cbdv.202300084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 11/29/2023]
Abstract
The fatty acid profile, antioxidant/antibacterial, and cytotoxic effects of the extracts obtained from Jurinea turcica B.Doğan& A.Duran have been evaluated for the first time in the current study. The fatty acid profile of ethanolic extracts was determined using the Soxhlet extractor by a gas chromatography-mass spectrometer. The antioxidant and antibacterial activities were measured by 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging and ferrous reduction tests and the disc diffusion technique. Additionally, the cytotoxicity and wound healing assays were performed on A549 cells. The highest amount of component in the leaf extract was docosanoic acid methyl ester, whereas abundant arachidonic acid methyl ester was mainly found in the flower extract. The IC50 values, the 50 % scavenging value for the DPPH radical, were 179.13 and 124.67 μg/mL for the leaf and flower extracts, respectively. IC50 values (the half-maximal inhibitory concentration) were 10.4 and 24.7 μg/mL for the flower and leaf extracts, respectively. The leaf extract showed more potent antibacterial activity on Enterococcus faecalis (17 mm) and Staphylococcus aureus (16 mm) bacteria than the flower extract. In conclusion, the extracts of J. turcica have anti-cancerogenic and antibacterial effects. Leaf extracts have antibacterial and anti-metastatic effects, while flower extracts show antioxidant, cytotoxic, and apoptotic properties in A549 cells.
Collapse
Affiliation(s)
- Alican Bahadır Semerci
- Sakarya University, Science Faculty, Department of Biology, 54187, Sakarya, Turkey
- Necmettin Erbakan University, Ereğli Vocational School of Health Services, 42310, Konya, Turkey
| | - Merve Yıldırım
- Istanbul University, Science Faculty, Department of Biology, Molecular Biology Division, Vezneciler, 34134, Istanbul, Turkey
| | - Füsun Oztay
- Istanbul University, Science Faculty, Department of Biology, Molecular Biology Division, Vezneciler, 34134, Istanbul, Turkey
| | - Mehmet Sagıroglu
- Sakarya University, Science Faculty, Department of Biology, 54187, Sakarya, Turkey
| | - Kenan Tunç
- Sakarya University, Science Faculty, Department of Biology, 54187, Sakarya, Turkey
| |
Collapse
|
14
|
Yao H, Liang Z, Wang W, Niu C. Integrative analyses of transcriptomes and metabolomes provide insight into salinity adaption in Bangia (Rhodaphyta). Int J Biol Macromol 2023; 253:127466. [PMID: 37875187 DOI: 10.1016/j.ijbiomac.2023.127466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/13/2023] [Accepted: 10/14/2023] [Indexed: 10/26/2023]
Abstract
The salinity of the external environment poses a serious threat to most land plants. Although seaweeds can adapt to this, intertidal species are subject to wide fluctuations in salinity, including hypo- and hyper-saline conditions. The red algal genus Bangiales is a typical example; it is one of the oldest eukaryotes with sexual reproduction and has successfully adapted to both marine and freshwater environments. However, there is a dearth of research focused on elucidating the mechanism by which marine Bangia (Bangia fuscopurpurea) adapts to hypo-salinity, as well as the mechanism by which freshwater Bangia (Bangia atropurpurea) adapts to hyper-salinity. The objective of this study is to employ third-generation full-length transcriptome data and untargeted metabolome data, to provide insights into the salinity adaptation mechanism of as well as the evolutionary relationship between both Bangia species. B. fuscopurpurea and B. atropurpurea exhibited 9112 and 8772 differentially expressed genes (DEGs), respectively, during various periods of hyper-saline condition. These genes were primarily enriched in secondary metabolites and energy-related metabolic pathways. Additionally, B. fuscopurpurea displayed 16,285 DEGs during different periods of hypo-saline condition, which were mainly enriched in metabolic pathways related to ion transport and membrane proteins. In the hyper- and hypo-saline adapt response processes of B. fuscopurpurea, a total of 303 transcription factors were identified, which belonged to 26 families. Among these, 85 and 142 differential transcription factors were identified, respectively, mainly belonging to the C2H2 and MYB family. Similarly, in the response process of B. atropurpurea to hyper-saline condition, a total of 317 transcription factors were identified, mainly belonging to 17 families. Among these, 121 differential transcription factors were identified, mainly belonging to the C2H2 and bZIP family. Furthermore, a correlation analysis was conducted to examine the relationship between the transcriptional and metabolic levels of both species under saline adaptation. The findings demonstrated that Bangia exhibits intricate adaptations to salinity, which involve swift regulation of its photosynthetic processes, alternations in membrane contents, and a robust anti-oxidation system to mitigate the effects of excess redox energy during exposure to varying salinity. Notably, the unsaturated fat and glutathione metabolic pathways were found to be significantly enriched in this context.
Collapse
Affiliation(s)
- Haiqin Yao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao 266071, China
| | - Zhourui Liang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, China
| | - Wenjun Wang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, China.
| | - Citong Niu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, No. 106 Nanjing Road, Qingdao 266071, China
| |
Collapse
|
15
|
Amer EI, Allam SR, Hassan AY, El-Fakharany EM, Agwa MM, Khattab SN, Sheta E, El-Faham MH. Can antibody conjugated nanomicelles alter the prospect of antibody targeted therapy against schistosomiasis mansoni? PLoS Negl Trop Dis 2023; 17:e0011776. [PMID: 38039267 PMCID: PMC10691730 DOI: 10.1371/journal.pntd.0011776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 11/07/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND CLA (conjugated linoleic acid)-mediated activation of the schistosome tegument-associated sphingomyelinase and consequent disruption of the outer membrane might allow host antibodies to access the apical membrane antigens. Here, we investigated a novel approach to enhance specific antibody delivery to concealed surface membrane antigens of Schistosoma mansoni utilising antibody-conjugated-CLA nanomicelle technology. METHODOLOGY/PRINCIPAL FINDINGS We invented and characterised an amphiphilic CLA-loaded whey protein co-polymer (CLA-W) as an IV injectable protein nanocarrier. Rabbit anti-Schistosoma mansoni infection (anti-SmI) and anti-Schistosoma mansoni alkaline phosphatase specific IgG antibodies were purified from rabbit sera and conjugated to the surface of CLA-W co-polymer to form antibody-conjugated-CLA-W nanomicelles (Ab-CLA-W). We investigated the schistosomicidal effects of CLA-W and Ab-CLA-W in a mouse model of Schistosoma mansoni against early and late stages of infection. Results showed that conjugation of nanomicelles with antibodies, namely anti-SmI, significantly enhanced the micelles' schistosomicidal and anti-pathology activities at both the schistosomula and adult worm stages of the infection resulting in 64.6%-89.9% reductions in worm number; 72.5-94% and 66.4-85.2% reductions in hepatic eggs and granulomas, respectively. Treatment induced overall improvement in liver histopathology, reducing granuloma size and fibrosis and significantly affecting egg viability. Indirect immunofluorescence confirmed CLA-W-mediated antigen exposure on the worm surface. Electron microscopy revealed extensive ultrastructural damage in worm tegument induced by anti-SmI-CLA-W. CONCLUSION/SIGNIFICANCE The novel antibody-targeted nano-sized CLA delivery system offers great promise for treatment of Schistosoma mansoni infection and control of its transmission. Our in vivo observations confirm an immune-mediated enhanced effect of the schistosomicidal action of CLA and hints at the prospect of nanotechnology-based immunotherapy, not only for schistosomiasis, but also for other parasitic infections in which chemotherapy has been shown to be immune-dependent. The results propose that the immunodominant reactivity of the anti-SmI serum, Schistosoma mansoni fructose biphosphate aldolase, SmFBPA, merits serious attention as a therapeutic and vaccine candidate.
Collapse
Affiliation(s)
- Eglal I. Amer
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Sonia R. Allam
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Aceel Y. Hassan
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Esmail M. El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria, Egypt
| | - Mona M. Agwa
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Giza, Egypt
| | - Sherine N. Khattab
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Eman Sheta
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Marwa H. El-Faham
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
16
|
Monge P, Astudillo AM, Pereira L, Balboa MA, Balsinde J. Dynamics of Docosahexaenoic Acid Utilization by Mouse Peritoneal Macrophages. Biomolecules 2023; 13:1635. [PMID: 38002317 PMCID: PMC10669016 DOI: 10.3390/biom13111635] [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: 10/19/2023] [Revised: 11/02/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
In this work, the incorporation of docosahexaenoic acid (DHA) in mouse resident peritoneal macrophages and its redistribution within the various phospholipid classes were investigated. Choline glycerophospholipids (PC) behaved as the major initial acceptors of DHA. Prolonged incubation with the fatty acid resulted in the transfer of DHA from PC to ethanolamine glycerophospholipids (PE), reflecting phospholipid remodeling. This process resulted in the cells containing similar amounts of DHA in PC and PE in the resting state. Mass spectrometry-based lipidomic analyses of phospholipid molecular species indicated a marked abundance of DHA in ether phospholipids. Stimulation of the macrophages with yeast-derived zymosan resulted in significant decreases in the levels of all DHA-containing PC and PI species; however, no PE or PS molecular species were found to decrease. In contrast, the levels of an unusual DHA-containing species, namely PI(20:4/22:6), which was barely present in resting cells, were found to markedly increase under zymosan stimulation. The levels of this phospholipid also significantly increased when the calcium-ionophore A23187 or platelet-activating factor were used instead of zymosan to stimulate the macrophages. The study of the route involved in the synthesis of PI(20:4/22:6) suggested that this species is produced through deacylation/reacylation reactions. These results define the increases in PI(20:4/22:6) as a novel lipid metabolic marker of mouse macrophage activation, and provide novel information to understand the regulation of phospholipid fatty acid turnover in activated macrophages.
Collapse
Affiliation(s)
- Patricia Monge
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain (A.M.A.); (M.A.B.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alma M. Astudillo
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain (A.M.A.); (M.A.B.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Pereira
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain (A.M.A.); (M.A.B.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María A. Balboa
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain (A.M.A.); (M.A.B.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jesús Balsinde
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain (A.M.A.); (M.A.B.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
17
|
Giedt MS, Thomalla JM, White RP, Johnson MR, Lai ZW, Tootle TL, Welte MA. Adipose triglyceride lipase promotes prostaglandin-dependent actin remodeling by regulating substrate release from lipid droplets. Development 2023; 150:dev201516. [PMID: 37306387 PMCID: PMC10281261 DOI: 10.1242/dev.201516] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
Lipid droplets (LDs), crucial regulators of lipid metabolism, accumulate during oocyte development. However, their roles in fertility remain largely unknown. During Drosophila oogenesis, LD accumulation coincides with the actin remodeling necessary for follicle development. Loss of the LD-associated Adipose Triglyceride Lipase (ATGL) disrupts both actin bundle formation and cortical actin integrity, an unusual phenotype also seen when the prostaglandin (PG) synthase Pxt is missing. Dominant genetic interactions and PG treatment of follicles indicate that ATGL acts upstream of Pxt to regulate actin remodeling. Our data suggest that ATGL releases arachidonic acid (AA) from LDs to serve as the substrate for PG synthesis. Lipidomic analysis detects AA-containing triglycerides in ovaries, and these are increased when ATGL is lost. High levels of exogenous AA block follicle development; this is enhanced by impairing LD formation and suppressed by reducing ATGL. Together, these data support the model that AA stored in LD triglycerides is released by ATGL to drive the production of PGs, which promote the actin remodeling necessary for follicle development. We speculate that this pathway is conserved across organisms to regulate oocyte development and promote fertility.
Collapse
Affiliation(s)
- Michelle S. Giedt
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | - Roger P. White
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Matthew R. Johnson
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Zon Weng Lai
- Harvard T.H. Chan Advanced Multi-omics Platform, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tina L. Tootle
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Michael A. Welte
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
18
|
Caputo MJ, Li W, Kendall SJ, Larsen A, Weigel KA, White HM. Liver and Muscle Transcriptomes Differ in Mid-Lactation Cows Divergent in Feed Efficiency in the Presence or Absence of Supplemental Rumen-Protected Choline. Metabolites 2023; 13:1023. [PMID: 37755303 PMCID: PMC10536747 DOI: 10.3390/metabo13091023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/28/2023] Open
Abstract
Improving dairy cow feed efficiency is critical to the sustainability and profitability of dairy production, yet the underlying mechanisms that contribute to individual cow variation in feed efficiency are not fully understood. The objectives of this study were to (1) identify genes and associated pathways that are altered in cows with high- or low-residual feed intake (RFI) using RNA sequencing, and (2) determine if rumen-protected choline supplementation during mid-lactation would influence performance or feed efficiency. Mid-lactation (134 ± 20 days in milk) multiparous Holstein cows were randomly assigned to either supplementation of 0 g/d supplementation (CTL; n = 32) or 30 g/d of a rumen-protected choline product (RPC; 13.2 g choline ion; n = 32; Balchem Corp., New Hampton, NY, USA). Residual feed intake was determined as dry matter intake regressed on milk energy output, days in milk, body weight change, metabolic body weight, and dietary treatment. The 12 cows with the highest RFI (low feed efficient; LE) and 12 cows with the lowest RFI (high feed efficient; HE), balanced by dietary treatment, were selected for blood, liver, and muscle analysis. No differences in production or feed efficiency were detected with RPC supplementation, although albumin was greater and arachidonic acid tended to be greater in RPC cows. Concentrations of β-hydroxybutyrate were greater in HE cows. Between HE and LE, 268 and 315 differentially expressed genes in liver and muscle tissue, respectively, were identified through RNA sequencing. Pathway analysis indicated differences in cell cycling, oxidative stress, and immunity in liver and differences in glucose and fatty acid pathways in muscle. The current work indicates that unique differences in liver and muscle post-absorptive nutrient metabolism contribute to sources of variation in feed efficiency and that differences in amino acid and fatty acid oxidation, cell cycling, and immune function should be further examined.
Collapse
Affiliation(s)
- Malia J. Caputo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (M.J.C.); (S.J.K.); (A.L.); (K.A.W.)
| | - Wenli Li
- United States Department of Agriculture-Agriculture Research Station, Madison, WI 53706, USA;
| | - Sophia J. Kendall
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (M.J.C.); (S.J.K.); (A.L.); (K.A.W.)
| | - Anna Larsen
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (M.J.C.); (S.J.K.); (A.L.); (K.A.W.)
- United States Department of Agriculture-Agriculture Research Station, Madison, WI 53706, USA;
| | - Kent A. Weigel
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (M.J.C.); (S.J.K.); (A.L.); (K.A.W.)
| | - Heather M. White
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (M.J.C.); (S.J.K.); (A.L.); (K.A.W.)
| |
Collapse
|
19
|
Zhang X, Wang B, Chen S, Fu Y. Protective effects of Typhonii Rhizoma in rheumatoid arthritis rats revealed by integrated metabolomics and network pharmacology. Biomed Chromatogr 2023; 37:e5683. [PMID: 37161606 DOI: 10.1002/bmc.5683] [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: 03/03/2023] [Revised: 04/14/2023] [Accepted: 05/06/2023] [Indexed: 05/11/2023]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with a 0.5% prevalence worldwide. Inflammation, periosteal proliferation and joint destruction are the main clinical symptoms of RA. Typhonii Rhizoma (TR) is the dry tuber of the Araceae plant Typhonium giganteum Engl, and possesses many uses such as dispelling obstructive wind-phlegm and relieving pain. It is used for the clinical treatment of arthromyodynia and RA. However, the mechanism of action remains unclear. In this study, we first evaluated the effects of TR in type II collagen-induced RA model rats. Secondly, in serum metabolomics, TR could ameliorate 11 potential metabolites in RA model rats and reversed RA through pentose and glucuronate interconversions, sphingolipid metabolism, glycerophospholipid metabolism and tryptophan metabolism. To further explore the mechanisms of TR, 40 chemical constituents were used to establish a component-target interaction network. Some key genes were verified by in vitro pharmacological tests by integrating the results from the network pharmacology and metabolomics. The verification results showed that the mechanisms of TR against RA may be related to the inhibition of the production of inflammatory cytokines and the expression and function of HIF1-α. This study serves as a theoretical basis for the treatment of RA with TR.
Collapse
Affiliation(s)
- Xinya Zhang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Biying Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Suiqing Chen
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment and Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yu Fu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Henan Lingrui Pharmaceutical Company, Xinyang, Henan, China
| |
Collapse
|
20
|
Witt A, Mateska I, Palladini A, Sinha A, Wölk M, Harauma A, Bechmann N, Pamporaki C, Dahl A, Rothe M, Kopaliani I, Adolf C, Riester A, Wielockx B, Bornstein SR, Kroiss M, Peitzsch M, Moriguchi T, Fedorova M, Grzybek M, Chavakis T, Mirtschink P, Alexaki VI. Fatty acid desaturase 2 determines the lipidomic landscape and steroidogenic function of the adrenal gland. SCIENCE ADVANCES 2023; 9:eadf6710. [PMID: 37478183 PMCID: PMC10361602 DOI: 10.1126/sciadv.adf6710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 06/16/2023] [Indexed: 07/23/2023]
Abstract
Corticosteroids regulate vital processes, including stress responses, systemic metabolism, and blood pressure. Here, we show that corticosteroid synthesis is related to the polyunsaturated fatty acid (PUFA) content of mitochondrial phospholipids in adrenocortical cells. Inhibition of the rate-limiting enzyme of PUFA synthesis, fatty acid desaturase 2 (FADS2), leads to perturbations in the mitochondrial lipidome and diminishes steroidogenesis. Consistently, the adrenocortical mitochondria of Fads2-/- mice fed a diet with low PUFA concentration are structurally impaired and corticoid levels are decreased. On the contrary, FADS2 expression is elevated in the adrenal cortex of obese mice, and plasma corticosterone is increased, which can be counteracted by dietary supplementation with the FADS2 inhibitor SC-26192 or icosapent ethyl, an eicosapentaenoic acid ethyl ester. In humans, FADS2 expression is elevated in aldosterone-producing adenomas compared to non-active adenomas or nontumorous adrenocortical tissue and correlates with expression of steroidogenic genes. Our data demonstrate that FADS2-mediated PUFA synthesis determines adrenocortical steroidogenesis in health and disease.
Collapse
Affiliation(s)
- Anke Witt
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Ivona Mateska
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Alessandra Palladini
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Centre Munich at the University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, 85764, Germany
| | - Anupam Sinha
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Akiko Harauma
- School of Life and Environmental Science, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa, 252-5201, Japan
| | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Christina Pamporaki
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Andreas Dahl
- DRESDEN-Concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, 01307, Germany
| | | | - Irakli Kopaliani
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Christian Adolf
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, Munich, 80336, Germany
| | - Anna Riester
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, Munich, 80336, Germany
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Stefan R. Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Matthias Kroiss
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, Munich, 80336, Germany
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, Wuerzburg, 97080, Germany
| | - Mirko Peitzsch
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Toru Moriguchi
- School of Life and Environmental Science, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa, 252-5201, Japan
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Michal Grzybek
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Centre Munich at the University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, 85764, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Peter Mirtschink
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| |
Collapse
|
21
|
Han S, Applewhite S, DeCata J, Jones S, Cummings J, Wang S. Arachidonic acid reverses cholesterol and zinc inhibition of human voltage-gated proton channels. J Biol Chem 2023:104918. [PMID: 37315791 PMCID: PMC10344949 DOI: 10.1016/j.jbc.2023.104918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/08/2023] [Accepted: 06/09/2023] [Indexed: 06/16/2023] Open
Abstract
Unlike other members of the voltage-gated ion channel superfamily, voltage-gated proton (Hv) channels are solely composed of voltage sensor domains without separate ion-conducting pores. Due to their unique dependence on both voltage and transmembrane pH gradients, Hv channels normally open to mediate proton efflux. Multiple cellular ligands were also found to regulate the function of Hv channels, including Zn2+, cholesterol, polyunsaturated arachidonic acid, and albumin. Our previous work showed that Zn2+ and cholesterol inhibit the human voltage-gated proton channel hHv1 by stabilizing its S4 segment at resting state conformations. Released from phospholipids by phospholipase A2 in cells upon infection or injury, arachidonic acid regulates the function of many ion channels, including hHv1. In the present work, we examined the effects of arachidonic acid on purified hHv1 channels using liposome flux assays and revealed underlying structural mechanisms using single-molecule Fluorescence Resonance Energy Transfer (smFRET). Our data indicated that arachidonic acid strongly activates hHv1 channels by promoting transitions of the S4 segment towards opening or 'pre-opening' conformations. Moreover, we found that arachidonic acid even activates hHv1 channels inhibited by Zn2+ and cholesterol, providing a biophysical mechanism to activate hHv1 channels in non-excitable cells upon infection or injury.
Collapse
Affiliation(s)
- Shuo Han
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - Sarah Applewhite
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - Jenna DeCata
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - Samuel Jones
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - John Cummings
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA
| | - Shizhen Wang
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, Kansas City, MO 64110 USA.
| |
Collapse
|
22
|
Chai X, Wen L, Song Y, He X, Yue J, Wu J, Chen X, Cai Z, Qi Z. DEHP exposure elevated cardiovascular risk in obese mice by disturbing the arachidonic acid metabolism of gut microbiota. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 875:162615. [PMID: 36878288 DOI: 10.1016/j.scitotenv.2023.162615] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
Phthalate esters (PAEs) are one of the significant classes of emerging contaminants that are increasingly detected in environmental and human samples. Nevertheless, the current toxicity studies rarely report how PAEs affect the cardiovascular system, especially in obese individuals. In this study, diet-induced obese mice and corresponding normal mice were exposed to di(2-ethylhexyl) phthalate (DEHP) by oral gavage at environmentally relevant concentrations and key characteristics of cardiovascular risk were examined. The 16S rRNA and high-resolution mass spectrometry were used to investigate the alterations in the gut microbial profile and metabolic homeostasis. The results indicated that the cardiovascular system of fat individuals was more susceptible to DEHP exposure than mice in the lean group. 16S rRNA-based profiling and correlation analysis collectively suggested DEHP-induced gut microbial remodeling in fed a high-fat diet mice, represented by the abundance of the genus Faecalibaculum. Using metagenomic approaches, Faecalibaculum rodentium was identified as the top-ranked candidate bacterium. Additionally, metabolomics data revealed that DEHP exposure altered the gut metabolic homeostasis of arachidonic acid (AA), which is associated with adverse cardiovascular events. Finally, cultures of Faecalibaculum rodentium were treated with AA in vitro to verify the role of Faecalibaculum rodentium in altering AA metabolism. Our findings provide novel insights into DEHP exposure induced cardiovascular damage in obese individuals and suggest that AA could be used as a potential modulator of gut microbiota to prevent related diseases.
Collapse
Affiliation(s)
- Xuyang Chai
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Luyao Wen
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Yuanyuan Song
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Xiaochong He
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Jingxian Yue
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, Guangdong, China
| | - Jianlin Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Xin Chen
- Center for Reproductive Medicine, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan 528300, Guangdong, China
| | - Zongwei Cai
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, Guangdong, China; State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China.
| | - Zenghua Qi
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou 510006, Guangdong, China.
| |
Collapse
|
23
|
Yang H, Rothenberger E, Zhao T, Fan W, Kelly A, Attaya A, Fan D, Panigrahy D, Deng J. Regulation of inflammation in cancer by dietary eicosanoids. Pharmacol Ther 2023:108455. [PMID: 37257760 DOI: 10.1016/j.pharmthera.2023.108455] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Cancer is a major burden of disease worldwide and increasing evidence shows that inflammation contributes to cancer development and progression. Eicosanoids are derived from dietary polyunsaturated fatty acids, such as arachidonic acid (AA), and are mainly produced by a series of enzymatic pathways that include cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P-450 epoxygenase (CYP). Eicosanoids consist of at least several hundred individual molecules and play important roles in the inflammatory response and inflammation-related cancers. SCOPE AND APPROACH Dietary sources of AA and biosynthesis of eicosanoids from AA through different metabolic pathways are summarized. The bioactivities of eicosanoids and their potential molecular mechanisms on inflammation and cancer are revealed. Additionally, current challenges and limitations in eicosanoid research on inflammation-related cancer are discussed. KEY FINDINGS AND CONCLUSIONS Dietary AA generates a large variety of eicosanoids, including prostaglandins, thromboxane A2, leukotrienes, cysteinyl leukotrienes, lipoxins, hydroxyeicosatetraenoic acids (HETEs), and epoxyeicosatrienoic acids (EETs). Eicosanoids exert different bioactivities and mechanisms involved in the inflammation and related cancer developments. A deeper understanding of eicosanoid biology may be advantageous in cancer treatment and help to define cellular targets for further therapeutic development.
Collapse
Affiliation(s)
- Haixia Yang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| | - Eva Rothenberger
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Tong Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Wendong Fan
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Abigail Kelly
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ahmed Attaya
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, China
| | - Dipak Panigrahy
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Jianjun Deng
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an 710069, China; State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
24
|
Hirata Y, Ferreri C, Yamada Y, Inoue A, Sansone A, Vetica F, Suzuki W, Takano S, Noguchi T, Matsuzawa A, Chatgilialoglu C. Geometrical isomerization of arachidonic acid during lipid peroxidation interferes with ferroptosis. Free Radic Biol Med 2023:S0891-5849(23)00461-6. [PMID: 37257700 DOI: 10.1016/j.freeradbiomed.2023.05.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/02/2023]
Abstract
Geometrical mono-trans isomers of arachidonic acid (mtAA) are endogenous products of free radical-induced cis-trans double bond isomerization occurring to natural fatty acids during cell metabolism, including lipid peroxidation (LPO). Very little is known about the functional roles of mtAA and in general on the effects of mono-trans isomers of polyunsaturated fatty acids (mtPUFA) in various types of programmed cell death, including ferroptosis. Using HT1080 and MEF cell cultures, supplemented with 20 μM PUFA (i.e., AA, EPA or DHA) and their mtPUFA congeners, ferroptosis occurred in the presence of RSL3 (a direct inhibitor of glutathione peroxidase 4) only with the PUFA in their natural cis configuration, whereas mtPUFA showed an anti-ferroptotic effect. By performing the fatty acid-based membrane lipidome analyses, substantial differences emerged in the membrane fatty acid remodeling of the two different cell fates. In particular, during ferroptosis mtPUFA formation and their incorporation, together with the enrichment of SFA, occurred. This opens new perspectives in the role of the membrane composition for a ferroptotic outcome. While pre-treatment with AA promoted cell death for treatment with H2O2 and RSL3, mtAA did not. Cell death by AA supplementation was suppressed also in the presence of either ferroptosis inhibitors, such as the lipophilic antioxidant ferrostatin-1, or NADPH oxidase (NOX) inhibitors, including diphenyleneiodonium chloride and apocynin. Our results confirm a more complex scenario for ferroptosis than actually believed. While LPO processes are active, the importance of environmental lipid levels, balance among SFA, MUFA and PUFA in lipid pools and formation of mtPUFA influence the membrane phospholipid turnover, with crucial effects in the occurrence of cell death by ferroptosis.
Collapse
Affiliation(s)
- Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aoba-ku, Aramaki, Sendai, Miyagi, 980-8578, Japan
| | - Carla Ferreri
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale Delle Ricerche, Via Piero Gobetti 101, 40129, Bologna, Italy
| | - Yuto Yamada
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aoba-ku, Aramaki, Sendai, Miyagi, 980-8578, Japan
| | - Aya Inoue
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aoba-ku, Aramaki, Sendai, Miyagi, 980-8578, Japan
| | - Anna Sansone
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale Delle Ricerche, Via Piero Gobetti 101, 40129, Bologna, Italy
| | - Fabrizio Vetica
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale Delle Ricerche, Via Piero Gobetti 101, 40129, Bologna, Italy
| | - Wakana Suzuki
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aoba-ku, Aramaki, Sendai, Miyagi, 980-8578, Japan
| | - Saya Takano
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aoba-ku, Aramaki, Sendai, Miyagi, 980-8578, Japan
| | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aoba-ku, Aramaki, Sendai, Miyagi, 980-8578, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aoba-ku, Aramaki, Sendai, Miyagi, 980-8578, Japan.
| | - Chryssostomos Chatgilialoglu
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale Delle Ricerche, Via Piero Gobetti 101, 40129, Bologna, Italy; Center for Advanced Technologies, Adam Mickiewicz University, 61-614, Poznan, Poland.
| |
Collapse
|
25
|
Stewart KS, Gonzales KAU, Yuan S, Tierney MT, Bonny AR, Yang Y, Infarinato NR, Cowley CJ, Levorse JM, Pasolli HA, Ghosh S, Rothlin CV, Fuchs E. Stem cells tightly regulate dead cell clearance to maintain tissue fitness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541773. [PMID: 37293114 PMCID: PMC10245816 DOI: 10.1101/2023.05.22.541773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Macrophages and dendritic cells have long been appreciated for their ability to migrate to and engulf dying cells and debris, including some of the billions of cells that are naturally eliminated from our body daily. However, a substantial number of these dying cells are cleared by 'non-professional phagocytes', local epithelial cells that are critical to organismal fitness. How non-professional phagocytes sense and digest nearby apoptotic corpses while still performing their normal tissue functions is unclear. Here, we explore the molecular mechanisms underlying their multifunctionality. Exploiting the cyclical bouts of tissue regeneration and degeneration during the hair cycle, we show that stem cells can transiently become non-professional phagocytes when confronted with dying cells. Adoption of this phagocytic state requires both local lipids produced by apoptotic corpses to activate RXRα, and tissue-specific retinoids for RARγ activation. This dual factor dependency enables tight regulation of the genes requisite to activate phagocytic apoptotic clearance. The tunable phagocytic program we describe here offers an effective mechanism to offset phagocytic duties against the primary stem cell function of replenishing differentiated cells to preserve tissue integrity during homeostasis. Our findings have broad implications for other non-motile stem or progenitor cells which experience cell death in an immune-privileged niche.
Collapse
Affiliation(s)
- Katherine S Stewart
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Kevin AU Gonzales
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Shaopeng Yuan
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Matthew T Tierney
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Alain R Bonny
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Yihao Yang
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Nicole R Infarinato
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Christopher J Cowley
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - John M Levorse
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Hilda Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY, USA
| | - Sourav Ghosh
- Departments of Neurology and Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Carla V Rothlin
- Departments of Immunobiology and Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| |
Collapse
|
26
|
Mehta A, Ratre YK, Soni VK, Shukla D, Sonkar SC, Kumar A, Vishvakarma NK. Orchestral role of lipid metabolic reprogramming in T-cell malignancy. Front Oncol 2023; 13:1122789. [PMID: 37256177 PMCID: PMC10226149 DOI: 10.3389/fonc.2023.1122789] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/12/2023] [Indexed: 06/01/2023] Open
Abstract
The immune function of normal T cells partially depends on the maneuvering of lipid metabolism through various stages and subsets. Interestingly, T-cell malignancies also reprogram their lipid metabolism to fulfill bioenergetic demand for rapid division. The rewiring of lipid metabolism in T-cell malignancies not only provides survival benefits but also contributes to their stemness, invasion, metastasis, and angiogenesis. Owing to distinctive lipid metabolic programming in T-cell cancer, quantitative, qualitative, and spatial enrichment of specific lipid molecules occur. The formation of lipid rafts rich in cholesterol confers physical strength and sustains survival signals. The accumulation of lipids through de novo synthesis and uptake of free lipids contribute to the bioenergetic reserve required for robust demand during migration and metastasis. Lipid storage in cells leads to the formation of specialized structures known as lipid droplets. The inimitable changes in fatty acid synthesis (FAS) and fatty acid oxidation (FAO) are in dynamic balance in T-cell malignancies. FAO fuels the molecular pumps causing chemoresistance, while FAS offers structural and signaling lipids for rapid division. Lipid metabolism in T-cell cancer provides molecules having immunosuppressive abilities. Moreover, the distinctive composition of membrane lipids has implications for immune evasion by malignant cells of T-cell origin. Lipid droplets and lipid rafts are contributors to maintaining hallmarks of cancer in malignancies of T cells. In preclinical settings, molecular targeting of lipid metabolism in T-cell cancer potentiates the antitumor immunity and chemotherapeutic response. Thus, the direct and adjunct benefit of lipid metabolic targeting is expected to improve the clinical management of T-cell malignancies.
Collapse
Affiliation(s)
- Arundhati Mehta
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| | - Yashwant Kumar Ratre
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| | | | - Dhananjay Shukla
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| | - Subhash C. Sonkar
- Multidisciplinary Research Unit, Maulana Azad Medical College, University of Delhi, New Delhi, India
| | - Ajay Kumar
- Department of Zoology, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
27
|
Li L, Tian Y. The role of metabolic reprogramming of tumor-associated macrophages in shaping the immunosuppressive tumor microenvironment. Biomed Pharmacother 2023; 161:114504. [PMID: 37002579 DOI: 10.1016/j.biopha.2023.114504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Macrophages are potent immune effector cells in innate immunity and exert dual-effects in the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) make up a significant portion of TME immune cells. Similar to M1/M2 macrophages, TAMs are also highly plastic, and their functions are regulated by cytokines, chemokines and other factors in the TME. The metabolic changes in TAMs are significantly associated with polarization towards a protumour or antitumour phenotype. The metabolites generated via TAM metabolic reprogramming in turn promote tumor progression and immune tolerance. In this review, we explore the metabolic reprogramming of TAMs in terms of energy, amino acid and fatty acid metabolism and the potential roles of these changes in immune suppression.
Collapse
Affiliation(s)
- Lunxu Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
28
|
Sorokin AV, Arnardottir H, Svirydava M, Ng Q, Baumer Y, Berg A, Pantoja CJ, Florida E, Teague HL, Yang ZH, Dagur PK, Powell-Wiley TM, Yu ZX, Playford MP, Remaley AT, Mehta NN. Comparison of the dietary omega-3 fatty acids impact on murine psoriasis-like skin inflammation and associated lipid dysfunction. J Nutr Biochem 2023; 117:109348. [PMID: 37044136 DOI: 10.1016/j.jnutbio.2023.109348] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023]
Abstract
Persistent skin inflammation and impaired resolution are the main contributors to psoriasis and associated cardiometabolic complications. Omega-3 polyunsaturated fatty acids (PUFAs), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are known to exert beneficial effects on inflammatory response and lipid function. However, a specific role of omega-3 PUFAs in psoriasis and accompanied pathologies are still a matter of debate. Here, we carried out a direct comparison between EPA and DHA 12 weeks diet intervention treatment of psoriasis-like skin inflammation in the K14-Rac1V12 mouse model. By utilizing sensitive techniques, we targeted EPA- and DHA-derived specialized pro-resolving lipid mediators and identified tightly connected signaling pathways by RNA sequencing. Treatment with experimental diets significantly decreased circulating pro-inflammatory cytokines and bioactive lipid mediators, altered psoriasis macrophage phenotypes and genes of lipid oxidation. The superficial role of these changes was related to DHA treatment and included increased levels of resolvin D5, protectin DX and maresin 2 in the skin. EPA treated mice had less pronounced effects but demonstrated a decreased skin accumulation of prostaglandin E2 and thromboxane B2. These results indicate that modulating psoriasis skin inflammation with the omega-3 PUFAs may have clinical significance and DHA treatment might be considered over EPA in this specific disease.
Collapse
Affiliation(s)
- Alexander V Sorokin
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Hildur Arnardottir
- Division of Cardiovascular Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institute, Sweden
| | - Maryia Svirydava
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Qimin Ng
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yvonne Baumer
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander Berg
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carla J Pantoja
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth Florida
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heather L Teague
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhi-Hong Yang
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Pradeep K Dagur
- Flow Cytometry Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tiffany M Powell-Wiley
- Social Determinants of Obesity and Cardiovascular Risk Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zu-Xi Yu
- Pathology Core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martin P Playford
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, Cardiovascular and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
29
|
Tallima H, El Ridi R. Mechanisms of Arachidonic Acid In Vitro Tumoricidal Impact. Molecules 2023; 28:molecules28041727. [PMID: 36838715 PMCID: PMC9966399 DOI: 10.3390/molecules28041727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/27/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
To promote the potential of arachidonic acid (ARA) for cancer prevention and management, experiments were implemented to disclose the mechanisms of its tumoricidal action. Hepatocellular, lung, and breast carcinoma and normal hepatocytes cell lines were exposed to 0 or 50 μM ARA for 30 min and then assessed for proliferative capacity, surface membrane-associated sphingomyelin (SM) content, neutral sphingomyelinase (nSMase) activity, beta 2 microglobulin (β2 m) expression, and ceramide (Cer) levels. Reactive oxygen species (ROS) content and caspase 3/7 activity were evaluated. Exposure to ARA for 30 min led to impairment of the tumor cells' proliferative capacity and revealed that the different cell lines display remarkably similar surface membrane SM content but diverse responses to ARA treatment. Arachidonic acid tumoricidal impact was shown to be associated with nSMase activation, exposure of cell surface membrane β2 m to antibody binding, and hydrolysis of SM to Cer, which accumulated on the cell surface and in the cytosol. The ARA and Cer-mediated inhibition of tumor cell viability appeared to be independent of ROS generation or caspase 3/7 activation. The data were compared and contrasted to findings reported in the literature on ARA tumoricidal mechanisms.
Collapse
Affiliation(s)
- Hatem Tallima
- Department of Chemistry, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
- Correspondence:
| | - Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
30
|
Toelzer C, Gupta K, Berger I, Schaffitzel C. Cryo-EM reveals binding of linoleic acid to SARS-CoV-2 spike glycoprotein, suggesting an antiviral treatment strategy. Acta Crystallogr D Struct Biol 2023; 79:111-121. [PMID: 36762857 PMCID: PMC9912919 DOI: 10.1107/s2059798323000049] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/03/2023] [Indexed: 01/21/2023] Open
Abstract
The COVID-19 pandemic and concomitant lockdowns presented a global health challenge and triggered unprecedented research efforts to elucidate the molecular mechanisms and pathogenicity of SARS-CoV-2. The spike glycoprotein decorating the surface of SARS-CoV-2 virions is a prime target for vaccine development, antibody therapy and serology as it binds the host cell receptor and is central for viral cell entry. The electron cryo-microscopy structure of the spike protein revealed a hydrophobic pocket in the receptor-binding domain that is occupied by an essential fatty acid, linoleic acid (LA). The LA-bound spike protein adopts a non-infectious locked conformation which is more stable than the infectious form and shields important immunogenic epitopes. Here, the impact of LA binding on viral infectivity and replication, and the evolutionary conservation of the pocket in other highly pathogenic coronaviruses, including SARS-CoV-2 variants of concern (VOCs), are reviewed. The importance of LA metabolic products, the eicosanoids, in regulating the human immune response and inflammation is highlighted. Lipid and fatty-acid binding to a hydrophobic pocket in proteins on the virion surface appears to be a broader strategy employed by viruses, including picornaviruses and Zika virus. Ligand binding stabilizes their protein structure and assembly, and downregulates infectivity. In the case of rhinoviruses, this has been exploited to develop small-molecule antiviral drugs that bind to the hydrophobic pocket. The results suggest a COVID-19 antiviral treatment based on the LA-binding pocket.
Collapse
Affiliation(s)
- Christine Toelzer
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, United Kingdom
- Bristol Synthetic Biology Centre: BrisSynBio, 24 Tyndall Avenue, Bristol BS8 1TQ, United Kingdom
| | - Kapil Gupta
- Imophoron Ltd, St Philips Central, Albert Road, Bristol BS2 0XJ, United Kingdom
| | - Imre Berger
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, United Kingdom
- Bristol Synthetic Biology Centre: BrisSynBio, 24 Tyndall Avenue, Bristol BS8 1TQ, United Kingdom
- Max Planck Bristol Centre for Minimal Biology, Cantock’s Close, Bristol BS8 1TS, United Kingdom
| | - Christiane Schaffitzel
- School of Biochemistry, University of Bristol, 1 Tankard’s Close, Bristol BS8 1TD, United Kingdom
- Bristol Synthetic Biology Centre: BrisSynBio, 24 Tyndall Avenue, Bristol BS8 1TQ, United Kingdom
| |
Collapse
|
31
|
Sanches JM, Zhao LN, Salehi A, Wollheim CB, Kaldis P. Pathophysiology of type 2 diabetes and the impact of altered metabolic interorgan crosstalk. FEBS J 2023; 290:620-648. [PMID: 34847289 DOI: 10.1111/febs.16306] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/14/2021] [Accepted: 11/29/2021] [Indexed: 02/06/2023]
Abstract
Diabetes is a complex and multifactorial disease that affects millions of people worldwide, reducing the quality of life significantly, and results in grave consequences for our health care system. In type 2 diabetes (T2D), the lack of β-cell compensatory mechanisms overcoming peripherally developed insulin resistance is a paramount factor leading to disturbed blood glucose levels and lipid metabolism. Impaired β-cell functions and insulin resistance have been studied extensively resulting in a good understanding of these pathways but much less is known about interorgan crosstalk, which we define as signaling between tissues by secreted factors. Besides hormones and organokines, dysregulated blood glucose and long-lasting hyperglycemia in T2D is associated with changes in metabolism with metabolites from different tissues contributing to the development of this disease. Recent data suggest that metabolites, such as lipids including free fatty acids and amino acids, play important roles in the interorgan crosstalk during the development of T2D. In general, metabolic remodeling affects physiological homeostasis and impacts the development of T2D. Hence, we highlight the importance of metabolic interorgan crosstalk in this review to gain enhanced knowledge of the pathophysiology of T2D, which may lead to new therapeutic approaches to treat this disease.
Collapse
Affiliation(s)
| | - Li Na Zhao
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Albert Salehi
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Claes B Wollheim
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| |
Collapse
|
32
|
Shu DY, Chaudhary S, Cho KS, Lennikov A, Miller WP, Thorn DC, Yang M, McKay TB. Role of Oxidative Stress in Ocular Diseases: A Balancing Act. Metabolites 2023; 13:187. [PMID: 36837806 PMCID: PMC9960073 DOI: 10.3390/metabo13020187] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Redox homeostasis is a delicate balancing act of maintaining appropriate levels of antioxidant defense mechanisms and reactive oxidizing oxygen and nitrogen species. Any disruption of this balance leads to oxidative stress, which is a key pathogenic factor in several ocular diseases. In this review, we present the current evidence for oxidative stress and mitochondrial dysfunction in conditions affecting both the anterior segment (e.g., dry eye disease, keratoconus, cataract) and posterior segment (age-related macular degeneration, proliferative vitreoretinopathy, diabetic retinopathy, glaucoma) of the human eye. We posit that further development of therapeutic interventions to promote pro-regenerative responses and maintenance of the redox balance may delay or prevent the progression of these major ocular pathologies. Continued efforts in this field will not only yield a better understanding of the molecular mechanisms underlying the pathogenesis of ocular diseases but also enable the identification of novel druggable redox targets and antioxidant therapies.
Collapse
Affiliation(s)
- Daisy Y. Shu
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Suman Chaudhary
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Kin-Sang Cho
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Anton Lennikov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - William P. Miller
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - David C. Thorn
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Menglu Yang
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Tina B. McKay
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
33
|
Roberts BM, Kolb AL, Geddis AV, Naimo MA, Matheny RW. The dose-response effects of arachidonic acid on primary human skeletal myoblasts and myotubes. J Int Soc Sports Nutr 2023; 20:2164209. [PMID: 36620755 PMCID: PMC9817121 DOI: 10.1080/15502783.2022.2164209] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Cellular inflammatory response, mediated by arachidonic acid (AA) and cyclooxygenase, is a highly regulated process that leads to the repair of damaged tissue. Recent studies on murine C2C12 cells have demonstrated that AA supplementation leads to myotube hypertrophy. However, AA has not been tested on primary human muscle cells. Therefore, the purpose of this study was to determine whether AA supplementation has similar effects on human muscle cells. Methods Proliferating and differentiating human myoblasts were exposed to AA in a dose-dependent manner (50-0.80 µM) for 48 (myoblasts) or 72 (myotubes) hours. Cell viability was tested using a 3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay and cell counting; myotube area was determined by immunocytochemistry and confocal microscopy; and anabolic signaling pathways were evaluated by western blot and RT-PCR. Results Our data show that the treatment of primary human myoblasts treated with 50 µM and 25 µM of AA led to the release of PGE2 and PGF2α at levels higher than those of control-treated cells (p < 0.001 for all concentrations). Additionally, 50 µM and 25 µM of AA suppressed myoblast proliferation, myotube area, and myotube fusion. Anabolic signaling indicated reductions in total and phosphorylated TSC2, AKT, S6, and 4EBP1 in myoblasts at 50 µM of AA (p < 0.01 for all), but not in myotubes. These changes were not affected by COX-2 inhibition with celecoxib. Conclusion Together, our data demonstrate that high concentrations of AA inhibit myoblast proliferation, myotube fusion, and myotube hypertrophy, thus revealing potential deleterious effects of AA on human skeletal muscle cell health and viability.
Collapse
Affiliation(s)
- Brandon M. Roberts
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA,CONTACT Brandon M. Roberts Military Performance Division, USARIEM10 General Greene Ave, Bldg. 42, Natick, MA
| | - Alexander L. Kolb
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Alyssa V. Geddis
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Marshall A. Naimo
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| | - Ronald W. Matheny
- Military Performance Division, US Army Research Institute of Environmental Medicine, Natick, MA, USA
| |
Collapse
|
34
|
Chen L, Zhao Q, Du X, Chen X, Jiao Q, Jiang H. Effects of oxidative stress caused by iron overload on arachidonic acid metabolites in MES23.5 cells. J Biosci 2022. [DOI: 10.1007/s12038-022-00321-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
35
|
Dey R, Dey S, Samadder A, Saxena AK, Nandi S. Natural Inhibitors against Potential Targets of Cyclooxygenase, Lipoxygenase and Leukotrienes. Comb Chem High Throughput Screen 2022; 25:2341-2357. [PMID: 34533441 DOI: 10.2174/1386207325666210917111847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/14/2021] [Accepted: 07/18/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND Cyclooxygenase (COX) and Lipoxygenase (LOX) enzymes catalyze the production of pain mediators like Prostaglandins (PGs) and Leukotrienes (LTs), respectively from arachidonic acid. INTRODUCTION The COX and LOX enzyme modulators are responsible for the major PGs and LTs mediated complications like asthma, osteoarthritis, rheumatoid arthritis, cancer, Alzheimer's disease, neuropathy and Cardiovascular Syndromes (CVS). Many synthetic Nonsteroidal Anti- Inflammatory Drugs (NSAIDs) used in the treatment have serious side effects like nausea, vomiting, hyperacidity, gastrointestinal ulcers, CVS, etc. Methods: The natural inhibitors of pain mediators have great acceptance worldwide due to fewer side effects on long-term uses. The present review is an extensive study of the advantages of plantbased vs synthetic inhibitors. RESULTS These natural COX and LOX inhibitors control inflammatory response without causing side-effect-related complicacy. CONCLUSION Therefore, the natural COX and LOX inhibitors may be used as alternative medicines for the management of pain and inflammation due to their less toxicity and resistivity.
Collapse
Affiliation(s)
- Rishita Dey
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Sudatta Dey
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Asmita Samadder
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Anil Kumar Saxena
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur-244713, India
| | - Sisir Nandi
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur-244713, India
| |
Collapse
|
36
|
Chen C, Wang W, Poklis JL, Lichtman AH, Ritter JK, Hu G, Xie D, Li N. Inactivation of fatty acid amide hydrolase protects against ischemic reperfusion injury-induced renal fibrogenesis. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166456. [PMID: 35710061 PMCID: PMC10215004 DOI: 10.1016/j.bbadis.2022.166456] [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: 04/20/2022] [Revised: 05/26/2022] [Accepted: 06/06/2022] [Indexed: 11/26/2022]
Abstract
Although cannabinoid receptors (CB) are recognized as targets for renal fibrosis, the roles of endogenous cannabinoid anandamide (AEA) and its primary hydrolytic enzyme, fatty acid amide hydrolase (FAAH), in renal fibrogenesis remain unclear. The present study used a mouse model of post-ischemia-reperfusion renal injury (PIR) to test the hypothesis that FAAH participates in the renal fibrogenesis. Our results demonstrated that PIR showed upregulated expression of FAAH in renal proximal tubules, accompanied with decreased AEA levels in kidneys. Faah knockout mice recovered the reduced AEA levels and ameliorated PIR-triggered increases in blood urea nitrogen, plasma creatinine as well as renal profibrogenic markers and injuries. Correspondingly, a selective FAAH inhibitor, PF-04457845, inhibited the transforming growth factor-beta 1 (TGF-β1)-induced profibrogenic markers in human proximal tubular cell line (HK-2 cells) and mouse primary cultured tubular cells. Knockdown of FAAH by siRNA in HK-2 cells had similar effects as PF-04457845. Tubular cells isolated from Faah-/- mice further validated the protection against TGF-β1-induced damages. The CB 1 or CB2 receptor antagonist and exogenous FAAH metabolite arachidonic acid failed to reverse the protective effects of FAAH inactivation in HK-2 cells. However, a substrate-selective inhibitor of AEA-cyclooxygenase-2 (COX-2) pathway significantly suppressed the anti-profibrogenic actions of FAAH inhibition. Further, the AEA-COX-2 metabolite, prostamide E2 exerted anti-fibrogenesis effect. These findings suggest that FAAH activation and the consequent reduction of AEA contribute to the renal fibrogenesis, and that FAAH inhibition protects against fibrogenesis in renal cells independently of CB receptors via the AEA-COX-2 pathway by the recovery of reduced AEA.
Collapse
Affiliation(s)
- Chaoling Chen
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Weili Wang
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Justin L Poklis
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Aron H Lichtman
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Joseph K Ritter
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Gaizun Hu
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Dengpiao Xie
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Ningjun Li
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
37
|
Wang K, Shi TQ, Lin L, Wei P, Ledesma-Amaro R, Ji XJ. Engineering Yarrowia lipolytica to Produce Tailored Chain-Length Fatty Acids and Their Derivatives. ACS Synth Biol 2022; 11:2564-2577. [DOI: 10.1021/acssynbio.2c00305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Kaifeng Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing 211816, People’s Republic of China
| | - Tian-Qiong Shi
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing 210046, People’s Republic of China
| | - Lu Lin
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing 211816, People’s Republic of China
| | - Ping Wei
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing 211816, People’s Republic of China
| | - Rodrigo Ledesma-Amaro
- Department of Bioengineering and Imperial College Centre for Synthetic Biology, Imperial College London, London SW7 2AZ, United Kindom
| | - Xiao-Jun Ji
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, No. 30 South Puzhu Road, Nanjing 211816, People’s Republic of China
| |
Collapse
|
38
|
Wan Y, Yang L, Li H, Ren H, Zhu K, Dong Z, Jiang S, Shang E, Qian D, Duan J. Zingiber officinale and Panax ginseng ameliorate ulcerative colitis in mice via modulating gut microbiota and its metabolites. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1203:123313. [PMID: 35662877 DOI: 10.1016/j.jchromb.2022.123313] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/19/2022] [Accepted: 05/26/2022] [Indexed: 12/17/2022]
Abstract
Zingiber officinale and Panax ginseng, as well-known traditional Chinese medicines, have been used together to clinically treat ulcerative colitis with synergistic effects for thousands of years. However, their compatibility mechanism remains unclear. In this study, the shift of gut microbiome and fecal metabolic profiles were monitored by 16S rRNA sequencing technology and ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry analysis, respectively, which aimed to reveal the synergistic mechanism of Zingiber officinale and Panax ginseng on the amelioration of ulcerative colitis. The results showed that the relative abundance of beneficial bacteria (such as Muribaculaceae_norank, Lachnospiraceae NK4A136 group and Akkermansia) was significantly increased and the abundance of pathogenic bacteria (such as Bacteroides, Parabacteroides and Desulfovibrio) was markedly decreased after the intervention of Zingiber officinale-Panax ginseng herb pair. And a total of 16 differential metabolites related to ulcerative colitis were identified by the metabolomics analysis, which were majorly associated with the metabolic pathways, including arachidonic acid metabolism, tryptophan metabolism, and steroid biosynthesis. Based on these findings, it was suggested that the regulation of the gut microbiota-metabolite axis might be a potential target for the synergistic mechanism of Zingiber officinale-Panax ginseng herb pair in the treatment of ulcerative colitis. Furthermore, the integrated analysis of microbiome and metabolomics used in this study could also serve as a useful template for exploring the mechanism of other drugs.
Collapse
Affiliation(s)
- Yue Wan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Lei Yang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Huifang Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Hui Ren
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Ke Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Zhiling Dong
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China.
| | - Erxin Shang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Dawei Qian
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China.
| |
Collapse
|
39
|
Novel Unspecific Peroxygenase from Truncatella angustata Catalyzes the Synthesis of Bioactive Lipid Mediators. Microorganisms 2022; 10:microorganisms10071267. [PMID: 35888989 PMCID: PMC9322767 DOI: 10.3390/microorganisms10071267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/10/2022] [Accepted: 06/16/2022] [Indexed: 02/01/2023] Open
Abstract
Lipid mediators, such as epoxidized or hydroxylated eicosanoids (EETs, HETEs) of arachidonic acid (AA), are important signaling molecules and play diverse roles at different physiological and pathophysiological levels. The EETs and HETEs formed by the cytochrome P450 enzymes are still not fully explored, but show interesting anti-inflammatory properties, which make them attractive as potential therapeutic target or even as therapeutic agents. Conventional methods of chemical synthesis require several steps and complex separation techniques and lead only to low yields. Using the newly discovered unspecific peroxygenase TanUPO from the ascomycetous fungus Truncatella angustata, 90% regioselective conversion of AA to 14,15-EET could be achieved. Selective conversion of AA to 18-HETE, 19-HETE as well as to 11,12-EET and 14,15-EET was also demonstrated with known peroxygenases, i.e., AaeUPO, CraUPO, MroUPO, MweUPO and CglUPO. The metabolites were confirmed by HPLC-ELSD, MS1 and MS2 spectrometry as well as by comparing their analytical data with authentic standards. Protein structure simulations of TanUPO provided insights into its substrate access channel and give an explanation for the selective oxyfunctionalization of AA. The present study expands the scope of UPOs as they can now be used for selective syntheses of AA metabolites that serve as reference material for diagnostics, for structure-function elucidation as well as for therapeutic and pharmacological purposes.
Collapse
|
40
|
Silonov SB, Kryvenko EO, Silonova NB, Shevchenko TM. The effect of vitamin E on the lipid environment of rat hepatocyte membranes. REGULATORY MECHANISMS IN BIOSYSTEMS 2022. [DOI: 10.15421/022213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Tocopherol is one of the known beneficial natural antioxidants ensuring the optimal level of functioning of mammalian organisms. Numerous in vitro and in vivo experiments have shown that the biological role of vitamin E is to prevent the development of pathologies caused by oxidative stress. In particular, the role of enzymatic factors of lipid peroxidation and related inflammation as a result of eicosanoid synthesis was clearly shown. We studied changes in the structural and functional state of hepatocyte membranes in the classical model of E-hypovitaminosis caused by long-term (70 days) insufficient intake of vitamin E in the diet of rats. The test components were determined spectrophotometrically after appropriate chromatographic procedures. The amount of total and individual leukotrienes was determined by ELISA. Prolonged tocopherol deficiency in rats caused a 49.4% decrease in tocopherol, more than 27.0% – in cholesterol. Of the 8 individual phospholipids studied, 6 showed significant changes: a decrease in cardiolipin and phosphatidylserine, and an increase in phosphatidylethanolamine by 3.24 times, an increse in lysophosphatidylcholine by 86.9%, in phosphatidylcholine by 52.8%, and in sphingomyelin by 30.6%, relative to control. There were changes in the levels of unsaturated fatty acids playing a significant role in the development of functional disorders in cells and affecting the metabolism of ecosanoids derived from arachidonic acid by the 5-lipoxygenase oxidation pathway. Changes in the levels of total and individual cysteinyl leukotrienes in the state of E-hypovitaminosis were revealed. Restoration of vitamin E intake returns most of the studied indicators such as tocopherol, cholesterol, polyunsaturated fatty acids to the control levels and activates the processes of sequential conversion of leukotrienes in the body of rats. The obtained results indicate the potentiating effect of vitamin E on metabolic processes in the body as a whole and in hepatocytes and eicosanoid metabolism. The degree of tocopherol intake allows one to influence the course of inflammatory processes associated with eicosanoids, not only through the impact on precursors, but also on the utilization of metabolites, including leukotrienes.
Collapse
|
41
|
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: 5.0] [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
|
42
|
Staufer O, Gupta K, Hernandez Bücher JE, Kohler F, Sigl C, Singh G, Vasileiou K, Yagüe Relimpio A, Macher M, Fabritz S, Dietz H, Cavalcanti Adam EA, Schaffitzel C, Ruggieri A, Platzman I, Berger I, Spatz JP. Synthetic virions reveal fatty acid-coupled adaptive immunogenicity of SARS-CoV-2 spike glycoprotein. Nat Commun 2022; 13:868. [PMID: 35165285 PMCID: PMC8844029 DOI: 10.1038/s41467-022-28446-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 infection is a major global public health concern with incompletely understood pathogenesis. The SARS-CoV-2 spike (S) glycoprotein comprises a highly conserved free fatty acid binding pocket (FABP) with unknown function and evolutionary selection advantage1,2. Deciphering FABP impact on COVID-19 progression is challenged by the heterogenous nature and large molecular variability of live virus. Here we create synthetic minimal virions (MiniVs) of wild-type and mutant SARS-CoV-2 with precise molecular composition and programmable complexity by bottom-up assembly. MiniV-based systematic assessment of S free fatty acid (FFA) binding reveals that FABP functions as an allosteric regulatory site enabling adaptation of SARS-CoV-2 immunogenicity to inflammation states via binding of pro-inflammatory FFAs. This is achieved by regulation of the S open-to-close equilibrium and the exposure of both, the receptor binding domain (RBD) and the SARS-CoV-2 RGD motif that is responsible for integrin co-receptor engagement. We find that the FDA-approved drugs vitamin K and dexamethasone modulate S-based cell binding in an FABP-like manner. In inflammatory FFA environments, neutralizing immunoglobulins from human convalescent COVID-19 donors lose neutralization activity. Empowered by our MiniV technology, we suggest a conserved mechanism by which SARS-CoV-2 dynamically couples its immunogenicity to the host immune response.
Collapse
Affiliation(s)
- Oskar Staufer
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany.
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany.
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany.
| | - Kapil Gupta
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, University of Bristol, 4 Tyndall Ave, Bristol, BS8 1TQ, UK
| | - Jochen Estebano Hernandez Bücher
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
| | - Fabian Kohler
- Department of Physics, Technical University of Munich, 85748, Garching, Germany
| | - Christian Sigl
- Department of Physics, Technical University of Munich, 85748, Garching, Germany
| | - Gunjita Singh
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
| | - Kate Vasileiou
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
| | - Ana Yagüe Relimpio
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
| | - Meline Macher
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Sebastian Fabritz
- Department for Chemical Biology, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Hendrik Dietz
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany
- Department of Physics, Technical University of Munich, 85748, Garching, Germany
| | - Elisabetta Ada Cavalcanti Adam
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Christiane Schaffitzel
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
- Bristol Synthetic Biology Centre BrisSynBio, University of Bristol, 4 Tyndall Ave, Bristol, BS8 1TQ, UK
- Halo Therapeutics Ltd, Science Creates, Albert Road St. Philips Central, Bristol, BS2 0XJ, UK
| | - Alessia Ruggieri
- Department of Infectious Diseases, Molecular Virology, Center for Integrated Infectious Disease Research, University of Heidelberg, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany
| | - Ilia Platzman
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK
| | - Imre Berger
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- School of Biochemistry, Biomedical Sciences, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- Bristol Synthetic Biology Centre BrisSynBio, University of Bristol, 4 Tyndall Ave, Bristol, BS8 1TQ, UK.
- Halo Therapeutics Ltd, Science Creates, Albert Road St. Philips Central, Bristol, BS2 0XJ, UK.
| | - Joachim P Spatz
- Department for Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany.
- Institute for Molecular Systems Engineering, University of Heidelberg, Im Neuenheimer Feld 253, 69120, Heidelberg, Germany.
- Max Planck-Bristol Center for Minimal Biology, University of Bristol, 1 Tankard's Close, Bristol, BS8 1TD, UK.
- Max Planck School Matter to Life, Jahnstraße 29, 69120, Heidelberg, Germany.
| |
Collapse
|
43
|
He T, Wang M, Kong J, Wang Q, Tian Y, Li C, Wang Q, Liu C, Huang J. Integrating network pharmacology and non-targeted metabolomics to explore the common mechanism of Coptis Categorized Formula improving T2DM zebrafish. JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114784. [PMID: 34718103 DOI: 10.1016/j.jep.2021.114784] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Coptis Categorized Formula (CCF) is one of the core prescriptions in Treatise on Febrile Diseases. Its efficacy can be available not only in exogenous diseases but widely in various internal injuries and miscellaneous diseases. CCF (i.e., Huanglian Jiedu Decoction, Huanglian Ejiao Decoction, Dahuang Huanglian Xiexin Decoction, Gegen Qinlian Decoction) is different in composition, but they all play a favorable role in curative effect on type 2 diabetes mellitus (T2DM). Therefore, it is of great significance to reveal the common mechanism of CCF in treating T2DM. AIM OF THE STUDY Based on network pharmacology and non-targeted metabolomics research strategy, the common mechanism of the CCF treating T2DM was discussed. MATERIALS AND METHODS Firstly, Ultra-high performance liquid chromatography-quadrupole-time of flight/mass spectrometry was used to identify the chemical constituents of the CCF. Then, the targets of these chemical components were used for network pharmacology analysis associated with therapeutic effect. Finally, the diabetic zebrafish model was constructed to further verify the common mechanism of the CCF in treating T2DM. RESULTS A total of 160 chemical compositions were identified and 16 of them were common chemical compositions of the four CCF, including berberine, baicalin, coptisine and so forth. Network pharmacology results showed that Dipeptidyl peptidase (DPP)-4, cysteinyl aspartate specific proteinase (CASP)3, nitric oxide synthase (NOS)2, NOS3, and other 37 targets were common targets of CCF, and advanced glycation end products (AGE)-receptor of advanced glycation end products (RAGE) signaling pathway in diabetic complications, mitogen-activated protein kinase (MAPK) signaling pathway and hypoxia inducible factor (HIF)-1 signaling pathway were critical pathways of four CCF in the treatment of T2DM. CCF can lessen the blood glucose of diabetic zebrafish. The contents of 25 differential metabolites in diabetic zebrafish were altered. These metabolites were mainly related to phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine metabolism, arachidonic acid metabolism, sphingolipid metabolism, and tyrosine metabolism. CONCLUSION Our research shows that the common mechanism of CCF in improving T2DM is as follows: berberine, baicalin, coptisine and other chemical components can directionally regulate DPP-4, CASP3, NOS2, NOS3 and other targets, which are mediated by AGE-RAGE signaling pathway in diabetic complications, MAPK signaling pathway and HIF-1 signaling pathway. The content of endogenous metabolites such as L-valine and L-sorbitose changes, and further regulates the metabolism of amino acid metabolism, lipid metabolism, purine metabolism, sphingosine metabolism and arachidonic acid metabolism, so as to play a significant role in regulating glycolipid metabolism, improving insulin resistance, inhibiting cell apoptosis, anti-oxidation and anti-inflammation, and finally ameliorating T2DM.
Collapse
Affiliation(s)
- Tao He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China
| | - Mingshuang Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing, 102488, China
| | - Jiao Kong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China
| | - Qiang Wang
- School of Pharmacy China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Yue Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China
| | - Chaofeng Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing, 102488, China
| | - Qian Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China
| | - Chuanxin Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China; Department of Metabolism and Endocrinology, Endocrine and Metabolic Disease Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology; Medical Key Laboratory of Hereditary Rare Diseases of Henan; Luoyang Sub-center of National Clinical Research Center for Metabolic Diseases, Luoyang, 471003, China.
| | - Jianmei Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Liangxiang Town, Fangshan District, Beijing, 102488, China.
| |
Collapse
|
44
|
Duttaroy AK, Basak S. Maternal Fatty Acid Metabolism in Pregnancy and Its Consequences in the Feto-Placental Development. Front Physiol 2022; 12:787848. [PMID: 35126178 PMCID: PMC8811195 DOI: 10.3389/fphys.2021.787848] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/30/2021] [Indexed: 12/16/2022] Open
Abstract
During pregnancy, maternal plasma fatty acids are critically required for cell growth and development, cell signaling, and the development of critical structural and functional aspects of the feto-placental unit. In addition, the fatty acids modulate the early stages of placental development by regulating angiogenesis in the first-trimester human placenta. Preferential transport of maternal plasma long-chain polyunsaturated fatty acids during the third trimester is critical for optimal fetal brain development. Maternal status such as obesity, diabetes, and dietary intakes may affect the functional changes in lipid metabolic processes in maternal-fetal lipid transport and metabolism. Fatty acids traverse the placental membranes via several plasma membrane fatty acid transport/binding proteins (FAT, FATP, p-FABPpm, and FFARs) and cytoplasmic fatty acid-binding proteins (FABPs). This review discusses the maternal metabolism of fatty acids and their effects on early placentation, placental fatty acid transport and metabolism, and their roles in feto-placental growth and development. The review also highlights how maternal fat metabolism modulates lipid processing, including transportation, esterification, and oxidation of fatty acids.
Collapse
Affiliation(s)
- Asim K. Duttaroy
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- *Correspondence: Asim K. Duttaroy,
| | - Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad, India
| |
Collapse
|
45
|
Ran X, Wang DW, Yu Z, Wu R, Zhang Q. Decreased Tissue Kallikrein Levels and the Risk of Ischemic Stroke: A Community-Based Cross-Sectional Study in China. J Inflamm Res 2022; 15:117-126. [PMID: 35023947 PMCID: PMC8747795 DOI: 10.2147/jir.s343972] [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: 10/12/2021] [Accepted: 12/22/2021] [Indexed: 11/23/2022] Open
Abstract
Aim Tissue kallikrein (TK) exerts protective effects on cardiac cerebrovascular diseases (CCVDs). Changes in TK level in plasma are associated with ischemic stroke and coronary artery disease (CAD); however, a causal correlation could not be established. Therefore, we investigated the association between TK levels and CCVDs in a community-based cross-sectional study in China. Methods A total of 6043 subjects (4242 men and 1801 women) were enrolled in this community-based cross-sectional study. Then, TK levels were measured using an enzyme-linked immunosorbent assay kit. Multivariate linear regression model and logistic regression were used to assess the correlations between TK levels and CCVDs. Subsequently, the receiver operating characteristic (ROC) curve was drawn to assess the value of TK level in evaluating the risk of ischemic stroke. Finally, the influence of various medications was evaluated on TK levels. Results The TK level was significantly lower in subjects with ischemic stroke (P < 0.001) and hypertension (P < 0.001) and negatively associated with ischemic stroke (P < 0.001) but not associated with hypertension, coronary heart disease, and diabetes compared to the traditional risk factors. The diagnostic accuracy for ischemic stroke, as quantified by the area under the curve, was 0.892 (95% CI, 0.884–0.900) for TK level, deeming it as a promising assessment tool. Moreover, no appreciable influence of various drugs therapy was found in TK levels (P = 0.222) except for those taking antilipemic agents. Conclusion TK is a strong and independent endogenous protective factor against ischemic stroke in the Chinese population and could be a promising biomarker for the risk of ischemic stroke.
Collapse
Affiliation(s)
- Xiao Ran
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Dao Wen Wang
- The Institute of Hypertension and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Zhen Yu
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Rongxue Wu
- Department of Biological Sciences Division/ Cardiology, University of Chicago, Chicago, IL, 60637, USA
| | - Qin Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| |
Collapse
|
46
|
Kim J, You S. Restoration of miR-223-3p expression in aged mouse uteri with Samul-tang administration. Integr Med Res 2022; 11:100835. [PMID: 35141134 PMCID: PMC8814392 DOI: 10.1016/j.imr.2022.100835] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 11/18/2022] Open
Abstract
Background Methods Results Conclusion
Collapse
Affiliation(s)
| | - Sooseong You
- Corresponding author at: Clinical Medicine Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Korea.
| |
Collapse
|
47
|
Reza AHMM, Zhu X, Qin J, Tang Y. Microalgae-Derived Health Supplements to Therapeutic Shifts: Redox-Based Study Opportunities with AIE-Based Technologies. Adv Healthc Mater 2021; 10:e2101223. [PMID: 34468087 DOI: 10.1002/adhm.202101223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/16/2021] [Indexed: 12/14/2022]
Abstract
Reactive oxygen species (ROS) are highly reactive molecules, serve the normal signaling in different cell types. Targeting ROS as the chemical signals, different stress based strategies have been developed to synthesis different anti-inflammatory molecules in microalgae. These molecules could be utilized as health supplements in human. To provoke the ROS-mediated defence systems, their connotation with the associated conditions must be well understood, therefore, proper tools for studying ROS in natural state are essential. The in vivo detection of ROS with phosphorescent probes offers promising opportunities to study these molecules in a non-invasive manner. Most of the common problems in the traditional fluorescent probes are lower photostability, excitation intensity, slow responsiveness, and the microenvironment that challenge their performance. Some ROS-specific aggregationinduced emission luminogens (AIEgens) with pronounced spatial and temporal resolution have recently demonstrated high selectivity, rapid responsiveness, and efficacies to resolve the aggregation-caused quenching issues. The nanocomposites of some AIE-photosensitizers can also improve the ROS-mediated photodynamic therapy. These AIEgens could be used to induce bioactive components in microalgae through altering the ROS signaling, therefore are more auspicious for biomedical research. This study reviews the prospects of AIEgen-based technologies to understand the ROS mediated bio-physiological processes in microalgae for better healthcare benefits.
Collapse
Affiliation(s)
- A. H. M. Mohsinul Reza
- College of Science and Engineering Flinders University South Australia 5042 Australia
- Institute for NanoScale Science and Technology Medical Device Research Institute College of Science and Engineering Flinders University South Australia 5042 Australia
| | - Xiaochen Zhu
- College of Science and Engineering Flinders University South Australia 5042 Australia
- Institute for NanoScale Science and Technology Medical Device Research Institute College of Science and Engineering Flinders University South Australia 5042 Australia
| | - Jianguang Qin
- College of Science and Engineering Flinders University South Australia 5042 Australia
| | - Youhong Tang
- College of Science and Engineering Flinders University South Australia 5042 Australia
- Institute for NanoScale Science and Technology Medical Device Research Institute College of Science and Engineering Flinders University South Australia 5042 Australia
| |
Collapse
|
48
|
Possible Interaction of Nonsteroidal Anti-inflammatory Drugs Against NF-κB- and COX-2-Mediated Inflammation: In Silico Probe. Appl Biochem Biotechnol 2021; 194:54-70. [PMID: 34843076 DOI: 10.1007/s12010-021-03719-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/08/2021] [Indexed: 10/19/2022]
Abstract
In recent years, inflammatory mediators have been considered a possible key for nonsteroidal anti-inflammatory drugs (NSAID's). NSAID's have been known as most promising medication against inflammation and its mediated pain. Inflammation could be recognize as a systemic adaptive stimulation triggered by detrimental stimuli as pathogenic attack and endogenous signals mediated injury inside the cells. In addition, there has been an inflammatory key mechanism involved in disease state. NSAIDs have been compromisingly recommended for targeting specific proteins and/or inflammatory-mediated enzymes including cyclooxygenases (COX). This subsequently inhibits the prostaglandins at the site of inflammation. For the past decades, two forms of the COX enzyme have been implicated as COX-1 expressed in cells and tissues and other COX-2 selectively triggered via proinflammatory cytokines at the site of inflammation and/or injury. In addition, NSAID's have also been implicated for the inhibition of NF-κB pathways, and other relevant proteins considered potent candidates for these drugs. NF-κB has been identified a classical proinflammatory signaling pathway. It has been recognized as a primary target for novel anti-inflammatory drugs. In our results, reports are being confirmed via the probable effects of NSAID's on inflammatory-mediated switches. Several studies were considered to enquire the possible interactions of NSAID's and inflammatory hub. Nevertheless, the exact mechanism is still debatable. In our study, NSAID's and their targeted proteins or molecules caused a convincing pattern. For improvised perception, the binding affinity of NSAID's with inflammatory-mediated proteins was quantified using a molecular docking tool. In addition, we have depicted the complex juncture of hydrogen bonding in targeted proteins with NSAID's. Our in silico investigations have revealed NSAID's as the powerful armor against COX-2- and NF-κB-mediated inflammation.
Collapse
|
49
|
Gerges SH, El-Kadi AOS. Sex differences in eicosanoid formation and metabolism: A possible mediator of sex discrepancies in cardiovascular diseases. Pharmacol Ther 2021; 234:108046. [PMID: 34808133 DOI: 10.1016/j.pharmthera.2021.108046] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022]
Abstract
Arachidonic acid is metabolized by cyclooxygenase, lipoxygenase, and cytochrome P450 enzymes to produce prostaglandins, leukotrienes, epoxyeicosatrienoic acids (EETs), and hydroxyeicosatetraenoic acids (HETEs), along with other eicosanoids. Eicosanoids have important physiological and pathological roles in the body, including the cardiovascular system. Evidence from several experimental and clinical studies indicates differences in eicosanoid levels, as well as in the activity or expression levels of their synthesizing and metabolizing enzymes between males and females. In addition, there is a clear state of gender specificity in cardiovascular diseases (CVD), which tend to be more common in men compared to women, and their risk increases significantly in postmenopausal women compared to younger women. This could be largely attributed to sex hormones, as androgens exert detrimental effects on the heart and blood vessels, whereas estrogen exhibits cardioprotective effects. Many of androgen and estrogen effects on the cardiovascular system are mediated by eicosanoids. For example, androgens increase the levels of cardiotoxic eicosanoids like 20-HETE, while estrogens increase the levels of cardioprotective EETs. Thus, sex differences in eicosanoid levels in the cardiovascular system could be an important underlying mechanism for the different effects of sex hormones and the differences in CVD between males and females. Understanding the role of eicosanoids in these differences can help improve the management of CVD.
Collapse
Affiliation(s)
- Samar H Gerges
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
50
|
Cioce M, Canino C, Pass H, Blandino G, Strano S, Fazio VM. Arachidonic acid drives adaptive responses to chemotherapy-induced stress in malignant mesothelioma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:344. [PMID: 34727953 PMCID: PMC8561918 DOI: 10.1186/s13046-021-02118-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Background High resistance to therapy and poor prognosis characterizes malignant pleural mesothelioma (MPM). In fact, the current lines of treatment, based on platinum and pemetrexed, have limited impact on the survival of MPM patients. Adaptive response to therapy-induced stress involves complex rearrangements of the MPM secretome, mediated by the acquisition of a senescence-associated-secretory-phenotype (SASP). This fuels the emergence of chemoresistant cell subpopulations, with specific gene expression traits and protumorigenic features. The SASP-driven rearrangement of MPM secretome takes days to weeks to occur. Thus, we have searched for early mediators of such adaptive process and focused on metabolites differentially released in mesothelioma vs mesothelial cell culture media, after treatment with pemetrexed. METHODS Mass spectrometry-based (LC/MS and GC/MS) identification of extracellular metabolites and unbiased statistical analysis were performed on the spent media of mesothelial and mesothelioma cell lines, at steady state and after a pulse with pharmacologically relevant doses of the drug. ELISA based evaluation of arachidonic acid (AA) levels and enzyme inhibition assays were used to explore the role of cPLA2 in AA release and that of LOX/COX-mediated processing of AA. QRT-PCR, flow cytometry analysis of ALDH expressing cells and 3D spheroid growth assays were employed to assess the role of AA at mediating chemoresistance features of MPM. ELISA based detection of p65 and IkBalpha were used to interrogate the NFkB pathway activation in AA-treated cells. RESULTS We first validated what is known or expected from the mechanism of action of the antifolate. Further, we found increased levels of PUFAs and, more specifically, arachidonic acid (AA), in the transformed cell lines treated with pemetrexed. We showed that pharmacologically relevant doses of AA tightly recapitulated the rearrangement of cell subpopulations and the gene expression changes happening in pemetrexed -treated cultures and related to chemoresistance. Further, we showed that release of AA following pemetrexed treatment was due to cPLA2 and that AA signaling impinged on NFkB activation and largely affected anchorage-independent, 3D growth and the resistance of the MPM 3D cultures to the drug. CONCLUSIONS AA is an early mediator of the adaptive response to pem in chemoresistant MPM and, possibly, other malignancies.
Collapse
Affiliation(s)
- Mario Cioce
- Department of Medicine, R.U. in Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128, Rome, Italy.
| | - Claudia Canino
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA.,Radiation Oncology Unit, UPMC Hillmann Cancer Center, San Pietro Hospital FBF, Rome, Italy
| | - Harvey Pass
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Sabrina Strano
- SAFU Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, 00144, Rome, Italy
| | - Vito Michele Fazio
- Department of Medicine, R.U. in Molecular Medicine and Biotechnology, University Campus Bio-Medico of Rome, 00128, Rome, Italy. .,Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133, Rome, Italy. .,Laboratory of Oncology, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013, San Giovanni Rotondo, Italy.
| |
Collapse
|