1
|
Wang Y, Ding S, Gao F, Jia Y, Wang X. Revealing the core active pharmaceutical ingredients and targets of Jie-gu capsules for fracture treatment through network pharmacology and mendelian randomization. Medicine (Baltimore) 2024; 103:e40798. [PMID: 39654220 PMCID: PMC11630937 DOI: 10.1097/md.0000000000040798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024] Open
Abstract
Jie-gu capsules are widely used for the treatment of fractures in China. However, the core active pharmaceutical ingredients of Jie-gu capsules and the potential mechanisms for treating fractures remain unclear. This study aims to preliminarily elucidate the potential mechanisms of Jie-gu capsules in the treatment of fractures through network pharmacology and mendelian randomization methods. Data of fracture patients were obtained from the GEO database (GSE93215), and the active pharmaceutical ingredients and therapeutic targets of Jie-gu capsules were retrieved from the TCMSP and TCMID databases to identify the intersection genes. Subsequently, a protein-protein interaction network of the intersection genes was constructed using the STRING database. Then, GO and KEGG analyses were conducted on the intersection genes. In addition, mendelian randomization was employed to identify core targets. Finally, molecular docking techniques were used to perform molecular docking of the core active pharmaceutical ingredients and core targets for Jie-gu capsules in the treatment of fractures. In this study, a total of 65 intersection genes involved in Jie-gu capsule treatment of fractures were identified. GO and KEGG results indicated that these 65 intersection genes were primarily associated with biological processes such as response to tumor necrosis factor and are involved in signaling pathways, especially the regulation of the MAPK signaling pathway. We identified 5 core active ingredients of Jie-gu capsules (quercetin, baicalein, kaempferol, luteolin, and succinic acid). Mendelian randomization confirmed 2 core targets (ALOX12 and EGF). Molecular docking results demonstrated that the core active pharmaceutical ingredients (quercetin, baicalein, kaempferol, luteolin, and succinic acid) exhibit high affinities with the core targets (ALOX12 and EGF). This study has unveiled the core active pharmaceutical ingredients and potential action targets of the Jie-gu capsules in treating fractures, offering valuable insights for subsequent foundational research and the development of new medications.
Collapse
Affiliation(s)
- Ying Wang
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Shuang Ding
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Feng Gao
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Yuyan Jia
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xukai Wang
- The Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| |
Collapse
|
2
|
Zhang B, Guo B, Kong H, Yang L, Yan H, Liu J, Zhou Y, An R, Wang F. Decoding the Ferroptosis-Related Gene Signatures and Immune Infiltration Patterns in Ovarian Cancer: Bioinformatic Prediction Integrated with Experimental Validation. J Inflamm Res 2024; 17:10333-10346. [PMID: 39654865 PMCID: PMC11626233 DOI: 10.2147/jir.s498740] [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: 09/30/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024] Open
Abstract
Background Ovarian cancer is a type of gynecological cancer with extremely high fatality rate. Ferroptosis, an iron-dependent regulated cell death, inhibits the immune infiltration of tumor cells. Therefore, it is worthwhile to explore the effects of ferroptosis-related gene signatures and immune infiltration patterns on the clinical prognosis of ovarian cancer. Methods In this study, we used the mRNA expression matrix and related medical information of those who suffer from ovarian cancer in the TCGA database. After that, we established a ferroptosis-related gene signature based on LASSO Cox regression model, and employed several specific enrichment analyses to explore the bioinformatics functions of differentially expressed genes (DEGs). Additionally, we analyzed the link between ferroptosis and immune cells by single-sample gene set enrichment analysis (ssGSEA) to create a heatmap of gene-immune cell correlation. We then examined the expression of immune checkpoints and verified the gene expression in ovarian cancer tissues by qPCR assays. Finally, we induced ferroptosis in ovarian cancer cells using drugs and analyzed their migration, invasion and gene expression. Results According to LASSO Cox regression analysis, 9 prognostic DEGs were in association with overall survival (OS), which was utilized to construct a 9-gene signature for patients. Patients were divided into two groups, in which high-risk group's OS was markedly shorter than that of low-risk group (Log-rank p<0.001). KEGG enrichment analysis showed that these DEGs were linked to human cytomegalovirus (HCMV) infection. The ssGSEA analysis revealed significant differences in immune cell type and expression between ALOX12 and GLRX5 groups (p<0.05). Heatmap showed high correlation of prognostic genes with various immune cells. qPCR assay confirmed the 9 gene expression signature in ovarian cancer tissues. The ovarian cancer cell invasion and migration were significantly inhibited after induction of ferroptosis. Conclusion We decoded the ferroptosis-related gene signatures and immune infiltration patterns that can be used to predict the prognosis of ovarian cancer patients.
Collapse
Affiliation(s)
- Beilei Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, People’s Republic of China
| | - Bin Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, People’s Republic of China
| | - Hancun Kong
- Department of Medical Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710004, People’s Republic of China
| | - Linwei Yang
- Department of Medical Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710004, People’s Republic of China
| | - Hui Yan
- Department of Medical Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710004, People’s Republic of China
| | - Jierui Liu
- Department of Medical Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710004, People’s Republic of China
| | - Yichen Zhou
- Department of Medical Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710004, People’s Republic of China
| | - Ruifang An
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, People’s Republic of China
| | - Fu Wang
- Department of Medical Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710004, People’s Republic of China
- Department of Biophysics, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, 710061, People’s Republic of China
- School of Pharmacy, Shaanxi University of International Trade & Commerce, Xianyang, Shaanxi, 712046, People’s Republic of China
| |
Collapse
|
3
|
Li X, Mao J. Research progress on the role of lipoxygenase and its inhibitors in prostate cancer. Future Oncol 2024; 20:3549-3568. [PMID: 39535136 PMCID: PMC11776861 DOI: 10.1080/14796694.2024.2419356] [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: 05/23/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Prostate cancer (PCa) has become a common disease among middle-aged and elderly men. The lipoxygenase (LOX) pathway plays a crucial role in the occurrence, development, invasion and metastasis of PCa and is therefore considered a new target for the prevention and treatment of PCa. 5-LOX and 12-LOX have a promoting effect on the occurrence, development, invasion and metastasis of PCa. 15-LOX-2 has an inhibitory effect on PCa. LOX inhibitors can effectively inhibit the metabolic activity of LOX. The research aims to review the mechanism of action and inhibitors of LOX in PCa, in order to provide relevant references for the prevention and treatment of PCa.
Collapse
Affiliation(s)
- Xiaobing Li
- Chongqing Medical & Pharmaceutical College, Chongqing, 400030, China
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Jingxin Mao
- Chongqing Medical & Pharmaceutical College, Chongqing, 400030, China
- College of Pharmaceutical Sciences, Southwest University, Chongqing400715, China
| |
Collapse
|
4
|
Olkowicz M, Karas A, Berkowicz P, Kaczara P, Jasztal A, Kurylowicz Z, Fedak F, Rosales-Solano H, Roy KS, Kij A, Buczek E, Pawliszyn J, Chlopicki S. Upregulation of ALOX12-12-HETE pathway impairs AMPK-dependent modulation of vascular metabolism in ApoE/LDLR -/- mice. Pharmacol Res 2024; 210:107478. [PMID: 39448044 DOI: 10.1016/j.phrs.2024.107478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Mitochondrial dysfunction and 12-lipoxygenase (ALOX12)-derived 12(S)-HETE production have been associated with vascular inflammation and the pathogenesis of atherosclerosis. However, the role of ALOX12 in regulating vascular energy metabolism in vascular inflammation has not been studied to date. Using mitochondrial and glycolysis functional profiling with the Seahorse extracellular flux analyzer, metabolipidomics, and proteomic analysis (LC-MS/MS), we characterized alterations in vascular energy metabolism in 2- and 6-month-old ApoE/LDLR-/- vs. control C57BL/6 mice. We identified that aorta of 6-month-old ApoE/LDLR-/- mice displayed compromised mitochondrial metabolism manifested by the reduced expression of mitochondrial enzymes, impaired mitochondrial respiration, and consequently diminished respiratory reserve capacity. An increased flux through the glycolysis/lactate shuttle, the hexosamine biosynthetic pathway (HBP), and the pentose phosphate pathway (PPP) was also recognized. Interestingly, ALOX12-12-HETE was the most upregulated axis in eicosanoid metabolism and histological examinations indicated that ApoE/LDLR-/- mice showed increased aortic expression of ALOX12, particularly in early atherosclerotic plaque areas. Remarkably, the joint blocking of ALOX12 and activation of AMPK, but not AMPK activation alone, resulted in the reprogramming of vascular metabolism, with improved mitochondrial respiration and suppressed auxiliary pathways (HBP, PPP, itaconate shunt). In conclusion, excessive activation of the ALOX12-12-HETE pathway in vascular inflammation in early atherosclerosis inhibits AMPK-dependent regulation of vascular metabolism. Consequently, ALOX12 may represent a novel target to boost impaired vascular mitochondrial function in pro-atherosclerotic vascular inflammation.
Collapse
Affiliation(s)
- Mariola Olkowicz
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland.
| | - Agnieszka Karas
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, Krakow 30-348, Poland
| | - Piotr Berkowicz
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Patrycja Kaczara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Zuzanna Kurylowicz
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Filip Fedak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, Krakow 30-348, Poland
| | - Hernando Rosales-Solano
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Kanchan Sinha Roy
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland
| | - Elzbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, Krakow 30-348, Poland
| | - Janusz Pawliszyn
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow 30-348, Poland; Department of Pharmacology, Jagiellonian University Medical College, Grzegorzecka 16, Krakow 31-531, Poland
| |
Collapse
|
5
|
Chikhaoui A, Zayoud K, Kraoua I, Bouchoucha S, Tebourbi A, Turki I, Yacoub-Youssef H. Supplementation with nicotinamide limits accelerated aging in affected individuals with cockayne syndrome and restores antioxidant defenses. Aging (Albany NY) 2024; 16:13271-13287. [PMID: 39611850 PMCID: PMC11719109 DOI: 10.18632/aging.206160] [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: 03/11/2024] [Accepted: 10/21/2024] [Indexed: 11/30/2024]
Abstract
Cockayne syndrome (CS) is a segmental progeroid syndrome characterized by defects in the DNA excision repair pathway, predisposing to neurodegenerative manifestations. It is a rare genetic disorder and an interesting model for studying premature aging. Oxidative stress and autophagy play an important role in the aging process. The study of these two processes in a model of accelerated aging and the means to counteract them would lead to the identification of relevant biomarkers with therapeutic value for healthy aging. Here we investigated the gene expression profiles of several oxidative stress-related transcripts derived from CS-affected individuals and healthy elderly donors. We also explored the effect of nicotinamide supplementation on several genes related to inflammation and autophagy. Gene expression analysis revealed alterations in two main pathways. This involves the activation of arachidonic acid metabolism and the repression of the NRF2 pathway in affected individuals with CS. The supplementation with nicotinamide adjusted these abnormalities by enhancing autophagy and decreasing inflammation. Furthermore, CSA/CSB-dependent depletion of the mitochondrial DNA polymerase-γ catalytic subunit (POLG1) was restored following nicotinamide supplementation in CS-affected individuals' fibroblasts. This study reveals the link between oxidative stress and accelerated aging in affected individuals with CS and highlights new biomarkers of cellular senescence. However, further analyses are needed to confirm these results, which could not be carried out, mainly due to the unavailability of crucial samples of this rare disease.
Collapse
Affiliation(s)
- Asma Chikhaoui
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| | - Kouloud Zayoud
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| | - Ichraf Kraoua
- Department of Neuropediatrics, National Institute of Neurology Mongi Ben Hamida, Tunis 2092, Tunisia
| | - Sami Bouchoucha
- Orthopedics Department, Béchir Hamza Children’s Hospital, Tunis 2092, Tunisia
| | - Anis Tebourbi
- Orthopedic and Trauma Surgery Department, Mongi Slim Hospital, La Marsa 2046, Tunisia
| | - Ilhem Turki
- Department of Neuropediatrics, National Institute of Neurology Mongi Ben Hamida, Tunis 2092, Tunisia
| | - Houda Yacoub-Youssef
- Laboratory of Biomedical Genomics and Oncogenetics, LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, El Manar I, Tunis 1002, Tunisia
| |
Collapse
|
6
|
Ağagündüz D, Yeşildemir Ö, Koçyiğit E, Koçak T, Özen Ünaldı B, Ayakdaş G, Budán F. Oxylipins Derived from PUFAs in Cardiometabolic Diseases: Mechanism of Actions and Possible Nutritional Interactions. Nutrients 2024; 16:3812. [PMID: 39599599 PMCID: PMC11597274 DOI: 10.3390/nu16223812] [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: 09/23/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Oxylipins are oxidized fatty acids, both saturated and unsaturated, formed through pathways that involve singlet oxygen or dioxygen-mediated oxygenation reactions and are primarily produced by enzyme families such as cyclooxygenases, lipoxygenases, and cytochrome P450. These lipid-based complex bioactive molecules are pivotal signal mediators, acting in a hormone-like manner in the pathophysiology of numerous diseases, especially cardiometabolic diseases via modulating plenty of mechanisms. It has been reported that omega-6 and omega-3 oxylipins are important novel biomarkers of cardiometabolic diseases. Moreover, collected literature has noted that diet and dietary components, especially fatty acids, can modulate these oxygenated lipid products since they are mainly derived from dietary omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) or linoleic acid and α-linolenic by elongation and desaturation pathways. This comprehensive review aims to examine their correlations to cardiometabolic diseases and how diets modulate oxylipins. Also, some aspects of developing new biomarkers and therapeutical utilization are detailed in this review.
Collapse
Affiliation(s)
- Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Türkiye
| | - Özge Yeşildemir
- Department of Nutrition and Dietetics, Bursa Uludag University, Görükle Campus, 16059 Bursa, Türkiye;
| | - Emine Koçyiğit
- Department of Nutrition and Dietetics, Ordu University, Cumhuriyet Yerleşkesi, 52200 Ordu, Türkiye;
| | - Tevfik Koçak
- Department of Nutrition and Dietetics, Gümüşhane University, Gümüşhanevî Kampüsü, 29100 Gümüşhane, Türkiye;
| | - Buket Özen Ünaldı
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Afyonkarahisar Health Sciences University, 03030 Afyonkarahisar, Türkiye;
| | - Gamze Ayakdaş
- Department of Nutrition and Dietetics, Acıbadem University, Kerem Aydınlar Campus, 34752 İstanbul, Türkiye;
| | - Ferenc Budán
- Institute of Physiology, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
7
|
Jaggernauth S, Ramoutar SR, Ramoutar R, Piaralal N. Is Papaya Leaf Extract the Sweet Remedy for Dengue-Induced Thrombocytopenia? Cureus 2024; 16:e71852. [PMID: 39559655 PMCID: PMC11571266 DOI: 10.7759/cureus.71852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2024] [Indexed: 11/20/2024] Open
Abstract
Dengue fever, caused by the dengue virus and transmitted by Aedes mosquitoes, poses a significant health challenge in tropical regions like Trinidad and Tobago, with severe cases leading to life-threatening complications. This case study presents a 65-year-old female with dengue exhibiting warning signs, characterized by persistent fever, abdominal pain, and thrombocytopenia. Despite supportive care, including intravenous fluids and symptomatic treatment, her platelet count remained critically low at 12.1 x 109/L. Following the introduction of diluted papaya leaf extract into her treatment regimen, her platelet count significantly improved to 122.7 x 109/L within two days, eventually returning to normal at follow-up appointments. This case suggests that Carica papaya leaf extract may provide therapeutic benefits in boosting platelet counts in dengue patients when used alongside conventional supportive care. Further research is essential to understand the extent of papaya's benefits and to integrate it safely into dengue management practices, potentially enhancing treatment options for affected individuals. In addition, public health efforts should be prioritized in raising awareness about dengue prevention, early diagnosis, and effective management strategies to reduce the disease's impact in affected regions.
Collapse
Affiliation(s)
- Sheneel Jaggernauth
- Department of Medicine, Eric Williams Medical Sciences Complex, Champ Fleurs, TTO
| | - Shyam R Ramoutar
- Department of Medicine, Eric Williams Medical Sciences Complex, Champ Fleurs, TTO
| | - Renuka Ramoutar
- Department of Opthalmology, Eric Williams Medical Sciences Complex, Champ Fleurs, TTO
| | - Neiala Piaralal
- Department of Medicine, Medical Associates Hospital, Champ Fleurs, TTO
| |
Collapse
|
8
|
Wickert A, Schwantes A, Fuhrmann DC, Brüne B. Inflammation in a ferroptotic environment. Front Pharmacol 2024; 15:1474285. [PMID: 39372215 PMCID: PMC11449703 DOI: 10.3389/fphar.2024.1474285] [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: 08/01/2024] [Accepted: 09/12/2024] [Indexed: 10/08/2024] Open
Abstract
Ferroptosis is an iron-dependent form of cell death, which finally culminates in lipid peroxidation and membrane damage. During the past decade, the interest in ferroptosis increased substantially and various regulatory components were discovered. The role of ferroptosis during inflammation and its impact on different immune cell populations is still under debate. Activation of inflammatory pathways such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and hypoxia inducible factors (HIFs) are known to alter the ability of cells to undergo ferroptosis and are closely connected to iron metabolism. During inflammation, iron regulatory systems fundamentally change and cells such as macrophages and neutrophils adapt their metabolism towards iron sequestering phenotypes. In this review, we discuss how ferroptosis alters inflammatory pathways and how iron metabolism under inflammatory conditions affects immune cell ferroptosis.
Collapse
Affiliation(s)
- Anja Wickert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Anna Schwantes
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Dominik C. Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
| |
Collapse
|
9
|
Zhou S, Hu J, Du S, Wang F, Fang Y, Zhang R, Wang Y, Zheng L, Gao M, Tang H. RNA-binding proteins potentially regulate alternative splicing of immune/inflammatory-associated genes during the progression of generalized pustular psoriasis. Arch Dermatol Res 2024; 316:538. [PMID: 39158708 DOI: 10.1007/s00403-024-03283-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 08/20/2024]
Abstract
Generalized pustular psoriasis (GPP) is a rare but severe form of psoriasis. However, the pathogenesis of GPP has not been fully elucidated. Although RNA-binding proteins (RBPs) and the alternative splicing (AS) process are essential for regulating post-transcriptional gene expression, their roles in GPP are still unclear. We aimed to elucidate the regulatory mechanisms to identify potential new therapeutic targets. Here, We analyzed an RNA sequencing (RNA-seq) dataset (GSE200977) of peripheral blood mononuclear cells (PBMCs) of 24 patients with GPP, psoriasis vulgaris (PV), and healthy controls (HCs) from the Gene Expression Omnibus (GEO) database. We found that the abnormal alternative splicing (AS) events associated with GPP were mainly "alt3p/alt5p", and 15 AS genes were differentially expressed. Notably, the proportions of different immune cell types were correlated with the expression levels of regulatory alternatively spliced genes (RASGs): significant differences were observed in expression levels of DTD2, NDUFAF3, NBPF15, and FBLN7 in B cells and ARFIP1, IPO11, and RP11-326L24.9 in neutrophils in the GPP samples. Furthermore, We identified 32 differentially expressed RNA-binding proteins (RBPs) (18 up-regulated and 14 down-regulated). Co-expression networks between 14 pairs of differentially expressed RBPs and RASGs were subsequently constructed, demonstrating that these differentially expressed RBPs may affect the progression of GPP by regulating the AS of downstream immune/inflammatory-related genes such as LINC00989, ENC1 and MMP25-AS1. Our results were innovative in revealing the involvement of inflammation-related RBPs and RASGs in the development of GPP from the perspective of RBP-regulated AS.
Collapse
Affiliation(s)
- Shan Zhou
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, 230032, Anhui, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Junjie Hu
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, 230032, Anhui, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Shuli Du
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, 230032, Anhui, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Fang Wang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, 230032, Anhui, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ying Fang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, 230032, Anhui, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ranran Zhang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, 230032, Anhui, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yixiao Wang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, 230032, Anhui, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Liyun Zheng
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China
- Institute of Dermatology, Anhui Medical University, Hefei, 230032, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, 230032, Anhui, China
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Min Gao
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China.
- Institute of Dermatology, Anhui Medical University, Hefei, 230032, Anhui, China.
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, 230032, Anhui, China.
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Huayang Tang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China.
- Institute of Dermatology, Anhui Medical University, Hefei, 230032, Anhui, China.
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, 230032, Anhui, China.
- Collaborative Innovation Center of Complex and Severe Skin Disease, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
10
|
Franks SJ, Gowler PRW, Dunster JL, Turnbull J, Gohir SA, Kelly A, Valdes AM, King JR, Barrett DA, Chapman V, Preston S. Modelling the role of enzymatic pathways in the metabolism of docosahexaenoic acid by monocytes and its association with osteoarthritic pain. Math Biosci 2024; 374:109228. [PMID: 38851528 DOI: 10.1016/j.mbs.2024.109228] [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: 02/09/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Chronic pain is a major cause of disability and suffering in osteoarthritis (OA) patients. Endogenous specialised pro-resolving molecules (SPMs) curtail pro-inflammatory responses. One of the SPM intermediate oxylipins, 17-hydroxydocasahexaenoic acid (17-HDHA, a metabolite of docosahexaenoic acid (DHA)), is significantly associated with OA pain. The aim of this multidisciplinary work is to develop a mathematical model to describe the contributions of enzymatic pathways (and the genes that encode them) to the metabolism of DHA by monocytes and to the levels of the down-stream metabolites, 17-HDHA and 14-hydroxydocasahexaenoic acid (14-HDHA), motivated by novel clinical data from a study involving 30 participants with OA. The data include measurements of oxylipin levels, mRNA levels, measures of OA severity and self-reported pain scores. We propose a system of ordinary differential equations to characterise associations between the different datasets, in order to determine the homeostatic concentrations of DHA, 17-HDHA and 14-HDHA, dependent upon the gene expression of the associated metabolic enzymes. Using parameter-fitting methods, local sensitivity and uncertainty analysis, the model is shown to fit well qualitatively to experimental data. The model suggests that up-regulation of some ALOX genes may lead to the down-regulation of 17-HDHA and that dosing with 17-HDHA increases the production of resolvins, which helps to down-regulate the inflammatory response. More generally, we explore the challenges and limitations of modelling real data, in particular individual variability, and also discuss the value of gathering additional experimental data motivated by the modelling insights.
Collapse
Affiliation(s)
- S J Franks
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK.
| | - P R W Gowler
- Pain Centre Versus Arthritis, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - J L Dunster
- Institute for Cardiovascular and Metabolic Research, University of Reading, UK
| | - J Turnbull
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK; NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK; Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - S A Gohir
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - A Kelly
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - A M Valdes
- Pain Centre Versus Arthritis, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK; NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - J R King
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| | - D A Barrett
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK; Centre for Analytical Bioscience, Advanced Materials and Healthcare Technologies Division, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - V Chapman
- Pain Centre Versus Arthritis, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK; School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK; NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - S Preston
- School of Mathematical Sciences, University of Nottingham, Nottingham, NG7 2RD, UK
| |
Collapse
|
11
|
Asai K, Lee HK, Sato S, Shimizu E, Jung J, Okazaki T, Ogawa M, Shimmura S, Tsubota K, Ogawa Y, Negishi K, Hirayama M. The Necroptosis Pathway Is Upregulated in the Cornea in Mice With Ocular Graft-Versus-Host Disease. Invest Ophthalmol Vis Sci 2024; 65:38. [PMID: 39189995 PMCID: PMC11361379 DOI: 10.1167/iovs.65.10.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/26/2024] [Indexed: 08/28/2024] Open
Abstract
Purpose To identify molecular signatures specific for ocular graft-versus-host disease (GVHD) by proteomic analysis of corneas from mice with GVHD. Methods We identified differentially expressed proteins (DEPs) in corneal samples from GVHD model mice and syngeneic control mice 4 weeks after bone marrow transplantation. Data-independent acquisition analysis was performed on individual samples, and the roles of DEPs in biological pathways related to GVHD were evaluated via bioinformatics and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Results Three important signaling pathways were upregulated in the cornea in mice with GVHD: (1) the necroptosis pathway, (2) the mitogen-activated protein kinase (MAPK) pathway, and (3) as previously reported, the neutrophil extracellular trap (NET) pathway. In those signaling pathways, we identified new upregulated molecules, including (1) receptor-interacting protein kinase 1 (RIPK1), RIPK3, interferon regulatory factor 9, the interferon-induced double-stranded RNA-activated protein kinase lipoxygenase, and high mobility group box1 (HMGB1) which are damage-associated molecular patterns (DAMPs) in the necroptosis pathway; (2) the sequentially upregulated interleukin 1 (IL-1) receptor-associated kinase (IRAK), an evolutionarily conserved signaling intermediate in the Toll pathway (ECSIT), and p38, which is downstream of the IL-1 receptor and increased CDC42/Rac (Rac2), a Rho family GTPase in the MAPK pathway; and (3) the integrin components CR3 and macrophage-1 antigen (MAC-1), which are DAMPs, and the pyroptosis-related protein gasdermin D (GSDMD) in the NET pathway. Conclusions These novel molecules may help researchers elucidate the pathogenesis of GVHD and identify new therapeutic targets for corneal changes in patients with ocular GVHD.
Collapse
Affiliation(s)
- Kazuki Asai
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Hyung Keun Lee
- Ophthalmology, Yonsei University College of Medicine, Seoul, South Korea
| | - Shinri Sato
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Eisuke Shimizu
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Jaehun Jung
- Ophthalmology, Yonsei University College of Medicine, Seoul, South Korea
| | - Takahiro Okazaki
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Mamoru Ogawa
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Shigeto Shimmura
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Department of Clinical Regenerative Medicine, Fujita Health University, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Tsubota Laboratory, Inc., Tokyo, Japan
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Masatoshi Hirayama
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
12
|
Siwicki MR, Kubes P. Varied roles for LGR6 in the immune response. Blood 2024; 144:352-354. [PMID: 39052274 DOI: 10.1182/blood.2024025017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
|
13
|
Wang X, Xin B, Guo M, Yu G, Wang J. GWASTool: A web pipeline for detecting SNP-phenotype associations. FUNDAMENTAL RESEARCH 2024; 4:761-769. [PMID: 39660349 PMCID: PMC11630686 DOI: 10.1016/j.fmre.2024.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/19/2024] [Accepted: 03/11/2024] [Indexed: 12/12/2024] Open
Abstract
The genome-wide association study (GWAS) aims to detect associations between individual single nucleotide polymorphisms (SNPs) or SNP interactions and phenotypes to decipher the genetic mechanism. Existing GWAS analysis tools have different focuses and advantages, but suffer a series of tedious and heterogeneous configurations for computation. It is inconvenient for researchers to simply choose and apply these tools, statistically and biologically analyze their results for different usages. To address these issues, we develop a user friendly web pipeline GWASTool for detecting associations, which includes simulation data generation, associated loci detection, result visualization, analysis and comparison. GWASTool provides a unified and plugin-able framework to encapsulate the heterogeneity of GWAS algorithms, simplifies the analysis steps and energizes GWAS tasks. GWASTool is implemented in Java and is freely available for public use at http://www.sdu-idea.cn/GWASTool. The website hosts a comprehensive collection of resources, including a user manual, description of integrated algorithms, data examples and standalone version for download.
Collapse
Affiliation(s)
- Xin Wang
- School of Software, Shandong University, Jinan 250101, China
- Joint SDU-NTU Centre for Artificial Intelligence Research, Shandong University, Jinan 250101, China
| | - Beibei Xin
- College of Agronomy & Biotechnology, China Agricultural University, Beijing 100193, China
| | - Maozu Guo
- College of Electrical and Information Engineering, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Guoxian Yu
- School of Software, Shandong University, Jinan 250101, China
- Joint SDU-NTU Centre for Artificial Intelligence Research, Shandong University, Jinan 250101, China
| | - Jun Wang
- Joint SDU-NTU Centre for Artificial Intelligence Research, Shandong University, Jinan 250101, China
| |
Collapse
|
14
|
Wang X, Peng W, Zhao Y, Sha J, Li N, Huang S, Wang H. Immune cell related signature predicts prognosis in esophageal squamous cell carcinoma based on single-cell and bulk-RNA sequencing. Front Oncol 2024; 14:1370801. [PMID: 38903709 PMCID: PMC11187079 DOI: 10.3389/fonc.2024.1370801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Background It has been reported that tumor immune microenvironment performs a vital role in tumor progress. However, acting mechanism of immune cell related genes (IRGs) in esophageal squamous cell carcinoma (ESCC) is uncertain. Methods TCGA-ESCC, GSE23400, GSE26886, GSE75241, and GSE196756 datasets were gained via public databases. First, differentially expressed genes (DEGs) between ESCC and control samples from GSE23400, GSE26886, and GSE75241 were screened out by differential expression analysis, and overlapping DEGs were identified. Single-cell transcriptome data of GSE196756 were applied to explore immune cells that might be involved in regulation of ESCC. Then, weighted gene co-expression network analysis was applied to screen IRGs. Next, differentially expressed IRGs (DE-IRGs) were identified by overlapping IRGs and DEGs, and were incorporated into univariate Cox, least absolute shrinkage and selection operator, and multivariate Cox to acquire prognosis-related genes, and ESCC samples were grouped into high-/low-risk groups on the basis of median risk score. Finally, the role of prognosis model in immunotherapy was analyzed. Results Totally 248 DEGs were yielded by overlapping 3,915 DEGs in GSE26886, 459 DEGs in GSE23400, and 1,641 DEGs in GSE75241. Single-cell analysis found that B cells, dendritic cells, monocytes, neutrophils, natural killer cells, and T cells were involved in ESCC development. Besides, MEred, MEblack, MEpink, MEblue and MEbrown modules were considered as key modules because of their highest correlations with immune cell subtypes. A total of 154 DE-IRGs were yielded by taking intersection of DEGs and genes in key modules. Moreover, CTSC, ALOX12, and RMND5B were identified as prognosis-related genes in ESCC. Obviously, Exclusion and TIDE scores were notably lower in high-risk group than in the other one, indicating that high-risk group was more responsive to immunotherapy. Conclusions Through bioinformatic analysis, we identified a prognosis model consisting of IRGs (CTSC, ALOX12, and RMND5B) in ESCC, providing new ideas for studies related to treatment and prognosis of ESCC.
Collapse
Affiliation(s)
- Xian Wang
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Wei Peng
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yali Zhao
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiming Sha
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Na Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shan Huang
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Pathology, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Gastroenterology, The Second People’s Hospital of Hefei, Hefei, China
| |
Collapse
|
15
|
Kern-Lunbery RJ, Rathert-Williams AR, Foote AP, Cunningham-Hollinger HC, Kuehn LA, Meyer AM, Lindholm-Perry AK. Genes involved in the cholecystokinin receptor signaling map were differentially expressed in the jejunum of steers with variation in residual feed intake. Vet Anim Sci 2024; 24:100357. [PMID: 38812584 PMCID: PMC11133974 DOI: 10.1016/j.vas.2024.100357] [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] [Indexed: 05/31/2024] Open
Abstract
The jejunum is a critical site for nutrient digestion and absorption, and variation in its ability to take up nutrients within the jejunum is likely to affect feed efficiency. The purpose of this study was to determine differences in gene expression in the jejunum of beef steers divergent for residual feed intake (RFI) in one cohort of steers (Year 1), and to validate those genes in animals from a second study (Year 2). Steers from Year 1 (n = 16) were selected for high and low RFI. Jejunum mucosal tissue was obtained for RNA-seq. Thirty-two genes were differentially expressed (PFDR≤0.15), and five were over-represented in pathways including inflammatory mediator, cholecystokinin receptor (CCKR) signaling, and p38 MAPK pathways. Several differentially expressed genes (ALOX12, ALPI, FABP6, FABP7, FLT1, GSTA2, MEF2B, PDK4, SPP1, and TTF2) have been previously associated with RFI in other studies. Real-time qPCR was used to validate nine differentially expressed genes in the Year 1 steers used for RNA-seq, and in the Year 2 validation cohort. Six genes were validated as differentially expressed (P < 0.1) using RT-qPCR in the Year 1 population. In the Year 2 population, five genes displayed the same direction of expression as the Year 1 population and 3 were differentially expressed (P < 0.1). The CCKR pathway is involved in digestion, appetite control, and regulation of body weight making it a compelling candidate for feed efficiency in cattle, and the validation of these genes in a second population of cattle is suggestive of a role in feed efficiency.
Collapse
Affiliation(s)
- Rebecca J. Kern-Lunbery
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933, USA
- Ward Laboratories, Inc., Kearney, NE 68848, USA
| | - Abigail R. Rathert-Williams
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933, USA
- University of Missouri, Division of Animal Sciences, Columbia, MO 65211, USA
| | - Andrew P. Foote
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933, USA
- Oklahoma State University, Department of Animal & Food Sciences, Stillwater, OK 74078, USA
| | | | - Larry A. Kuehn
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE 68933, USA
| | - Allison M. Meyer
- University of Missouri, Division of Animal Sciences, Columbia, MO 65211, USA
- University of Wyoming, Department of Animal Science, Laramie, WY 82071, USA
| | | |
Collapse
|
16
|
Xia Q, Gao W, Yang J, Xing Z, Ji Z. The deregulation of arachidonic acid metabolism in ovarian cancer. Front Oncol 2024; 14:1381894. [PMID: 38764576 PMCID: PMC11100328 DOI: 10.3389/fonc.2024.1381894] [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: 02/04/2024] [Accepted: 04/19/2024] [Indexed: 05/21/2024] Open
Abstract
Arachidonic acid (AA) is a crucial polyunsaturated fatty acid in the human body, metabolized through the pathways of COX, LOX, and cytochrome P450 oxidase to generate various metabolites. Recent studies have indicated that AA and its metabolites play significant regulatory roles in the onset and progression of ovarian cancer. This article examines the recent research advancements on the correlation between AA metabolites and ovarian cancer, both domestically and internationally, suggesting their potential use as biological markers for early diagnosis, targeted therapy, and prognosis monitoring.
Collapse
Affiliation(s)
- Qiuyi Xia
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Wen Gao
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jintao Yang
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhifang Xing
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhaodong Ji
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Xu X, Yu D, Wang Y, Xu P, Jiang X, Lu F, Liu S. Integrating network pharmacology and renal metabonomics to reveal the protective mechanism of resveratrol on gouty nephropathy. Biomed Chromatogr 2024; 38:e5839. [PMID: 38402638 DOI: 10.1002/bmc.5839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 02/27/2024]
Abstract
Resveratrol (Res) has been demonstrated to have beneficial effects on gouty nephropathy (GN). However, the mechanisms of Res on GN remain unclear. This study aimed to investigate the mechanisms of Res on GN. In this study, network pharmacology technology was used to predict the Res targets in the prevention and treatment of GN. Renal metabonomics was used to identify differential metabolites in kidney tissue of GN model rats. Finally, molecular docking technology was used to verify the binding ability of Res to key targets. Metabonomics analysis showed that 24 potentially important metabolites were involved in the prevention and treatment of GN with Res. After exposure to Res, metabolite levels normalized. The network pharmacology analysis showed that 24 key targets were involved in the prevention and treatment of GN disease. According to the metabolite-gene network diagram, we identified two core genes, PTGS1 and PTGS2, and found that both were involved in the arachidonic acid metabolism pathway. Molecular docking further verified the affinity of Res binding to PTGS1 and PTGS2. In conclusion, the mechanism of Res against GN may be the regulation of arachidonic acid metabolism through the regulation of PTGS 1 and PTGS 2.
Collapse
Affiliation(s)
- Xiaomin Xu
- Research Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Donghua Yu
- Research Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Yu Wang
- Research Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Peng Xu
- Research Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Xin Jiang
- Research Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Fang Lu
- Research Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Shumin Liu
- Research Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| |
Collapse
|
18
|
Matsumoto T, Murakami Y, Yoshida-Sakai N, Katsuchi D, Kanazawa K, Okamura T, Imamura Y, Ono M, Kuwano M. Enhanced ALOX12 Gene Expression Predicts Therapeutic Susceptibility to 5-Azacytidine in Patients with Myelodysplastic Syndromes. Int J Mol Sci 2024; 25:4583. [PMID: 38731802 PMCID: PMC11083213 DOI: 10.3390/ijms25094583] [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: 03/28/2024] [Revised: 04/17/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
5-azacytidine (AZA), a representative DNA-demethylating drug, has been widely used to treat myelodysplastic syndromes (MDS). However, it remains unclear whether AZA's DNA demethylation of any specific gene is correlated with clinical responses to AZA. In this study, we investigated genes that could contribute to the development of evidence-based epigenetic therapeutics with AZA. A DNA microarray identified that AZA specifically upregulated the expression of 438 genes in AZA-sensitive MDS-L cells but not in AZA-resistant counterpart MDS-L/CDA cells. Of these 438 genes, the ALOX12 gene was hypermethylated in MDS-L cells but not in MDS-L/CDA cells. In addition, we further found that (1) the ALOX12 gene was hypermethylated in patients with MDS compared to healthy controls; (2) MDS classes with excess blasts showed a relatively lower expression of ALOX12 than other classes; (3) a lower expression of ALOX12 correlated with higher bone marrow blasts and a shorter survival in patients with MDS; and (4) an increased ALOX12 expression after AZA treatment was associated with a favorable response to AZA treatment. Taking these factors together, an enhanced expression of the ALOX12 gene may predict favorable therapeutic responses to AZA therapy in MDS.
Collapse
Affiliation(s)
- Taichi Matsumoto
- Basic Medical Research Unit, St. Mary’s Research Center, 422, Tsubuku-Honmachi, Kurume 850-8543, Fukuoka, Japan; (Y.M.); (D.K.); (K.K.); (M.O.); (M.K.)
| | - Yuichi Murakami
- Basic Medical Research Unit, St. Mary’s Research Center, 422, Tsubuku-Honmachi, Kurume 850-8543, Fukuoka, Japan; (Y.M.); (D.K.); (K.K.); (M.O.); (M.K.)
| | - Nao Yoshida-Sakai
- Department of Hematology, St. Mary’s Hospital, 422, Tsubuku-Honmachi, Kurume 850-8543, Fukuoka, Japan; (N.Y.-S.); (T.O.); (Y.I.)
| | - Daisuke Katsuchi
- Basic Medical Research Unit, St. Mary’s Research Center, 422, Tsubuku-Honmachi, Kurume 850-8543, Fukuoka, Japan; (Y.M.); (D.K.); (K.K.); (M.O.); (M.K.)
| | - Kuon Kanazawa
- Basic Medical Research Unit, St. Mary’s Research Center, 422, Tsubuku-Honmachi, Kurume 850-8543, Fukuoka, Japan; (Y.M.); (D.K.); (K.K.); (M.O.); (M.K.)
| | - Takashi Okamura
- Department of Hematology, St. Mary’s Hospital, 422, Tsubuku-Honmachi, Kurume 850-8543, Fukuoka, Japan; (N.Y.-S.); (T.O.); (Y.I.)
| | - Yutaka Imamura
- Department of Hematology, St. Mary’s Hospital, 422, Tsubuku-Honmachi, Kurume 850-8543, Fukuoka, Japan; (N.Y.-S.); (T.O.); (Y.I.)
| | - Mayumi Ono
- Basic Medical Research Unit, St. Mary’s Research Center, 422, Tsubuku-Honmachi, Kurume 850-8543, Fukuoka, Japan; (Y.M.); (D.K.); (K.K.); (M.O.); (M.K.)
| | - Michihiko Kuwano
- Basic Medical Research Unit, St. Mary’s Research Center, 422, Tsubuku-Honmachi, Kurume 850-8543, Fukuoka, Japan; (Y.M.); (D.K.); (K.K.); (M.O.); (M.K.)
| |
Collapse
|
19
|
Meng YW, Liu JY. Pathological and pharmacological functions of the metabolites of polyunsaturated fatty acids mediated by cyclooxygenases, lipoxygenases, and cytochrome P450s in cancers. Pharmacol Ther 2024; 256:108612. [PMID: 38369063 DOI: 10.1016/j.pharmthera.2024.108612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/19/2024] [Accepted: 02/05/2024] [Indexed: 02/20/2024]
Abstract
Oxylipins have garnered increasing attention because they were consistently shown to play pathological and/or pharmacological roles in the development of multiple cancers. Oxylipins are the metabolites of polyunsaturated fatty acids via both enzymatic and nonenzymatic pathways. The enzymes mediating the metabolism of PUFAs include but not limited to lipoxygenases (LOXs), cyclooxygenases (COXs), and cytochrome P450s (CYPs) pathways, as well as the down-stream enzymes. Here, we systematically summarized the pleiotropic effects of oxylipins in different cancers through pathological and pharmacological aspects, with specific reference to the enzyme-mediated oxylipins. We discussed the specific roles of oxylipins on cancer onset, growth, invasion, and metastasis, as well as the expression changes in the associated metabolic enzymes and the associated underlying mechanisms. In addition, we also discussed the clinical application and potential of oxylipins and related metabolic enzymes as the targets for cancer prevention and treatment. We found the specific function of most oxylipins in cancers, especially the underlying mechanisms and clinic applications, deserves and needs further investigation. We believe that research on oxylipins will provide not only more therapeutic targets for various cancers but also dietary guidance for both cancer patients and healthy humans.
Collapse
Affiliation(s)
- Yi-Wen Meng
- CNTTI of the Institute of Life Sciences & Department of Anesthesia of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing 400016, China
| | - Jun-Yan Liu
- CNTTI of the Institute of Life Sciences & Department of Anesthesia of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing 400016, China; College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
20
|
Li XJ, Suo P, Wang YN, Zou L, Nie XL, Zhao YY, Miao H. Arachidonic acid metabolism as a therapeutic target in AKI-to-CKD transition. Front Pharmacol 2024; 15:1365802. [PMID: 38523633 PMCID: PMC10957658 DOI: 10.3389/fphar.2024.1365802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 03/26/2024] Open
Abstract
Arachidonic acid (AA) is a main component of cell membrane lipids. AA is mainly metabolized by three enzymes: cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (CYP450). Esterified AA is hydrolysed by phospholipase A2 into a free form that is further metabolized by COX, LOX and CYP450 to a wide range of bioactive mediators, including prostaglandins, lipoxins, thromboxanes, leukotrienes, hydroxyeicosatetraenoic acids and epoxyeicosatrienoic acids. Increased mitochondrial oxidative stress is considered to be a central mechanism in the pathophysiology of the kidney. Along with increased oxidative stress, apoptosis, inflammation and tissue fibrosis drive the progressive loss of kidney function, affecting the glomerular filtration barrier and the tubulointerstitium. Recent studies have shown that AA and its active derivative eicosanoids play important roles in the regulation of physiological kidney function and the pathogenesis of kidney disease. These factors are potentially novel biomarkers, especially in the context of their involvement in inflammatory processes and oxidative stress. In this review, we introduce the three main metabolic pathways of AA and discuss the molecular mechanisms by which these pathways affect the progression of acute kidney injury (AKI), diabetic nephropathy (DN) and renal cell carcinoma (RCC). This review may provide new therapeutic targets for the identification of AKI to CKD continuum.
Collapse
Affiliation(s)
- Xiao-Jun Li
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ping Suo
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yan-Ni Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Liang Zou
- School of Food and Bioengineering, Chengdu University, Chengdu, Sichuan, China
| | - Xiao-Li Nie
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying-Yong Zhao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Hua Miao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
21
|
Sun S, Peng K, Yang B, Yang M, Jia X, Wang N, Zhang Q, Kong D, Du Y. The therapeutic effect of wine-processed Corni Fructus on chronic renal failure in rats through the interference with the LPS/IL-1-mediated inhibition of RXR function. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117511. [PMID: 38036016 DOI: 10.1016/j.jep.2023.117511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Corni Fructus, derived from the fruit of Cornus officinalis Sieb. et Zucc, is a widely utilized traditional Chinese medicine (TCM) with established efficacy in the treatment of diverse chronic kidney diseases. Crude Corni Fructus (CCF) and wine-processed Corni Fructus (WCF) are the main processed forms of Corni Fructus. Generally, TCM is often used after processing (paozhi). Despite the extensive use of processed TCM, the underlying mechanisms of processing for most TCMs have been unclear so far. AIM OF THE STUDY In this study, an integrated strategy combined renal metabolomics with proteomics was established and investigated the potential processing mechanisms of CCF or WCF on chronic renal failure (CRF) models. MATERIALS AND METHODS Firstly, the differences in biochemical parameters and pathological histology were compared to evaluate the effects of CCF and WCF on CRF model rats. Then, the tissue differential metabolites and proteins between CCF and WCF on CRF model rats were screened based on metabolomics and proteomics technology. Concurrently, a combined approach of metabolomics and proteomics was employed to investigate the underlying mechanisms associated with these marker metabolic products and proteins. RESULTS Compared to the MG group, there were 27 distinct metabolites and 143 different proteins observed in the CCF-treatment group, while the WCF-treatment group exhibited 24 distinct metabolites and 379 different proteins. Further, the integration interactions analysis of the protein and lipid metabolite revealed that both WCF and CCF improved tryptophan degradation and LPS/IL-1-mediated inhibition of RXR function. WCF inhibited RXR function more than CCF via the modulation of LPS/IL-1 in the CRF model. Experimental results were validated by qRT-PCR and western blotting. Notably, the gene expression amount and protein levels of FMO3 and CYP2E1 among 8 genes influenced by WCF were higher compared to CCF. CONCLUSION The results of this study provide a theoretical basis for further study of Corni Fructus with different processing techniques in CRF. The findings also offer guidance for investigating the mechanism of action of herbal medicines in diseases employing diverse processing techniques.
Collapse
Affiliation(s)
- Shilin Sun
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China; Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Hebei, 071000, PR China
| | - Kenan Peng
- Hebei General Hospital, Shijiazhuang, Hebei, 050051, PR China
| | - Bingkun Yang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Mengxin Yang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Xinming Jia
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Nan Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Qian Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China
| | - Dezhi Kong
- Institute of Chinese Integrative Medicine, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China.
| | - Yingfeng Du
- Department of Pharmaceutical Analysis, School of Pharmacy, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang, Hebei, 050017, PR China.
| |
Collapse
|
22
|
Oliw EH. Thirty years with three-dimensional structures of lipoxygenases. Arch Biochem Biophys 2024; 752:109874. [PMID: 38145834 DOI: 10.1016/j.abb.2023.109874] [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: 09/29/2023] [Revised: 12/17/2023] [Accepted: 12/17/2023] [Indexed: 12/27/2023]
Abstract
The X-ray crystal structures of soybean lipoxygenase (LOX) and rabbit 15-LOX were reported in the 1990s. Subsequent 3D structures demonstrated a conserved U-like shape of the substrate cavities as reviewed here. The 8-LOX:arachidonic acid (AA) complex showed AA bound to the substrate cavity carboxylate-out with C10 at 3.4 Å from the iron metal center. A recent cryo-electron microscopy (EM) analysis of the 12-LOX:AA complex illustrated AA in the same position as in the 8-LOX:AA complex. The 15- and 12-LOX complexes with isoenzyme-specific inhibitors/substrate mimics confirmed the U-fold. 5-LOX oxidizes AA to leukotriene A4, the first step in biosynthesis of mediators of asthma. The X-ray structure showed that the entrance to the substrate cavity was closed to AA by Phe and Tyr residues of a partly unfolded α2-helix. Recent X-ray analysis revealed that soaking with inhibitors shifted the short α2-helix to a long and continuous, which opened the substrate cavity. The α2-helix also adopted two conformations in 15-LOX. 12-LOX dimers consisted of one closed and one open subunit with an elongated α2-helix. 13C-ENDOR-MD computations of the 9-MnLOX:linoleate complex showed carboxylate-out position with C11 placed 3.4 ± 0.1 Å from the catalytic water. 3D structures have provided a solid ground for future research.
Collapse
Affiliation(s)
- Ernst H Oliw
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE 751 24, Uppsala, Sweden.
| |
Collapse
|
23
|
Broos JY, van der Burgt RTM, Konings J, Rijnsburger M, Werz O, de Vries HE, Giera M, Kooij G. Arachidonic acid-derived lipid mediators in multiple sclerosis pathogenesis: fueling or dampening disease progression? J Neuroinflammation 2024; 21:21. [PMID: 38233951 PMCID: PMC10792915 DOI: 10.1186/s12974-023-02981-w] [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: 09/22/2023] [Accepted: 11/30/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS), characterized by neuroinflammation, demyelination, and neurodegeneration. Considering the increasing prevalence among young adults worldwide and the disabling phenotype of the disease, a deeper understanding of the complexity of the disease pathogenesis is needed to ultimately improve diagnosis and personalize treatment opportunities. Recent findings suggest that bioactive lipid mediators (LM) derived from ω-3/-6 polyunsaturated fatty acids (PUFA), also termed eicosanoids, may contribute to MS pathogenesis. For example, disturbances in LM profiles and especially those derived from the ω-6 PUFA arachidonic acid (AA) have been reported in people with MS (PwMS), where they may contribute to the chronicity of neuroinflammatory processes. Moreover, we have previously shown that certain AA-derived LMs also associated with neurodegenerative processes in PwMS, suggesting that AA-derived LMs are involved in more pathological events than solely neuroinflammation. Yet, to date, a comprehensive overview of the contribution of these LMs to MS-associated pathological processes remains elusive. MAIN BODY This review summarizes and critically evaluates the current body of literature on the eicosanoid biosynthetic pathway and its contribution to key pathological hallmarks of MS during different disease stages. Various parts of the eicosanoid pathway are highlighted, namely, the prostanoid, leukotriene, and hydroxyeicosatetraenoic acids (HETEs) biochemical routes that include specific enzymes of the cyclooxygenases (COXs) and lipoxygenases (LOX) families. In addition, cellular sources of LMs and their potential target cells based on receptor expression profiles will be discussed in the context of MS. Finally, we propose novel therapeutic approaches based on eicosanoid pathway and/or receptor modulation to ultimately target chronic neuroinflammation, demyelination and neurodegeneration in MS. SHORT CONCLUSION The eicosanoid pathway is intrinsically linked to specific aspects of MS pathogenesis. Therefore, we propose that novel intervention strategies, with the aim of accurately modulating the eicosanoid pathway towards the biosynthesis of beneficial LMs, can potentially contribute to more patient- and MS subtype-specific treatment opportunities to combat MS.
Collapse
Affiliation(s)
- Jelle Y Broos
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Rianne T M van der Burgt
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
| | - Julia Konings
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands
| | - Merel Rijnsburger
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Gijs Kooij
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC, location VU Medical Center, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Amsterdam UMC, Amsterdam, The Netherlands.
| |
Collapse
|
24
|
Ma Y, Cai G, Chen J, Yang X, Hua G, Han D, Li X, Feng D, Deng X. Combined transcriptome and metabolome analysis reveals breed-specific regulatory mechanisms in Dorper and Tan sheep. BMC Genomics 2024; 25:70. [PMID: 38233814 PMCID: PMC10795462 DOI: 10.1186/s12864-023-09870-9] [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: 06/05/2023] [Accepted: 12/04/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Dorper and Tan sheep are renowned for their rapid growth and exceptional meat quality, respectively. Previous research has provided evidence of the impact of gut microbiota on breed characteristics. The precise correlation between the gastrointestinal tract and peripheral organs in each breed is still unclear. Investigating the metabolic network of the intestinal organ has the potential to improve animal growth performance and enhance economic benefits through the regulation of intestinal metabolites. RESULTS In this study, we identified the growth advantage of Dorper sheep and the high fat content of Tan sheep. A transcriptome study of the brain, liver, skeletal muscle, and intestinal tissues of both breeds revealed 3,750 differentially expressed genes (DEGs). The genes PPARGC1A, LPL, and PHGDH were found to be highly expressed in Doper, resulting in the up-regulation of pathways related to lipid oxidation, glycerophospholipid metabolism, and amino acid anabolism. Tan sheep highly express the BSEP, LDLR, and ACHE genes, which up-regulate the pathways involved in bile transport and cholesterol homeostasis. Hindgut content analysis identified 200 differentially accumulated metabolites (DAMs). Purines, pyrimidines, bile acids, and fatty acid substances were more abundant in Dorper sheep. Based on combined gene and metabolite analyses, we have identified glycine, serine, and threonine metabolism, tryptophan metabolism, bile secretion, cholesterol metabolism, and neuroactive ligand-receptor interaction as key factors contributing to the differences among the breeds. CONCLUSIONS This study indicates that different breeds of sheep exhibit unique breed characteristics through various physiological regulatory methods. Dorper sheep upregulate metabolic signals related to glycine, serine, and threonine, resulting in an increase in purine and pyrimidine substances. This, in turn, promotes the synthesis of amino acids and facilitates body development, resulting in a faster rate of weight gain. Tan sheep accelerate bile transport, reduce bile accumulation in the intestine, and upregulate cholesterol homeostasis signals in skeletal muscles. This promotes the accumulation of peripheral and intramuscular fat, resulting in improved meat quality. This work adopts a joint analysis method of multi-tissue transcriptome and gut metabolome, providing a successful case for analyzing the mechanisms underlying the formation of various traits.
Collapse
Affiliation(s)
- Yuhao Ma
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Ganxian Cai
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Jianfei Chen
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Xue Yang
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Guoying Hua
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Deping Han
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China
| | - Xinhai Li
- Department of Animal Science and college of Agriculture, Ningxia University, Yinchuan, 750021, China
| | - Dengzhen Feng
- Department of Animal Science and college of Agriculture, Ningxia University, Yinchuan, 750021, China
| | - Xuemei Deng
- Key Laboratory of Animal Genetics, Breeding, and Reproduction of the Ministry of Agriculture and Beijing Key Laboratory of Animal Genetic Improvement, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
25
|
Zahoor I, Waters J, Ata N, Datta I, Pedersen TL, Cerghet M, Poisson L, Markovic-Plese S, Rattan R, Taha AY, Newman JW, Giri S. Blood-based targeted metabolipidomics reveals altered omega fatty acid-derived lipid mediators in relapsing-remitting multiple sclerosis patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.04.574253. [PMID: 38260401 PMCID: PMC10802284 DOI: 10.1101/2024.01.04.574253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Unresolved and uncontrolled inflammation is considered a hallmark of pathogenesis in chronic inflammatory diseases like multiple sclerosis (MS), suggesting a defective resolution process. Inflammatory resolution is an active process partially mediated by endogenous metabolites of dietary polyunsaturated fatty acids (PUFA), collectively termed specialized pro-resolving lipid mediators (SPMs). Altered levels of resolution mediators have been reported in several inflammatory diseases and may partly explain impaired inflammatory resolution. Performing LC-MS/MS-based targeted lipid mediator profiling, we observed distinct changes in fatty acid metabolites in serum from 30 relapsing-remitting MS (RRMS) patients relative to 30 matched healthy subjects (HS). Robust linear regression revealed 12 altered lipid mediators after adjusting for confounders (p <0.05). Of these, 15d-PGJ2, PGE3, and LTB5 were increased in MS while PGF2a, 8,9-DiHETrE, 5,6-DiHETrE, 20-HETE, 15-HETE, 12-HETE, 12-HEPE, 14-HDoHE, and DHEA were decreased in MS compared to HS. In addition, 12,13-DiHOME and 12,13-DiHODE were positively correlated with expanded disability status scale values (EDSS). Using Partial Least Squares, we identified several lipid mediators with high VIP scores (VIP > 1: 32% - 52%) of which POEA, PGE3, DHEA, LTB5, and 12-HETE were top predictors for distinguishing between RRMS and HS (AUC =0.75) based on the XGBoost Classifier algorithm. Collectively, these findings suggest an imbalance between inflammation and resolution. Altogether, lipid mediators appear to have potential as diagnostic and prognostic biomarkers for RRMS.
Collapse
Affiliation(s)
- Insha Zahoor
- Department of Neurology, Henry Ford Health, Detroit, 48202, USA
| | - Jeffrey Waters
- Department of Neurology, Henry Ford Health, Detroit, 48202, USA
| | - Nasar Ata
- Department of Neurology, Henry Ford Health, Detroit, 48202, USA
| | - Indrani Datta
- Department of Public Health Sciences, Henry Ford Health, Detroit, 48202, USA
| | | | - Mirela Cerghet
- Department of Neurology, Henry Ford Health, Detroit, 48202, USA
| | - Laila Poisson
- Department of Public Health Sciences, Henry Ford Health, Detroit, 48202, USA
| | - Silva Markovic-Plese
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ramandeep Rattan
- Division of Gynaecology Oncology, Department of Women’s Health Services, Henry Ford Health, Detroit, 48202, USA
| | - Ameer Y. Taha
- Department of Food and Technology, University of California, Davis, USA
- West Coast Metabolomics Center, Genome Center, University of California-Davis, Davis, CA, 95616, USA
| | - John W. Newman
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, 95616, USA
- West Coast Metabolomics Center, Genome Center, University of California-Davis, Davis, CA, 95616, USA
- Department of Nutrition, University of California-Davis, Davis, CA, 95616, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health, Detroit, 48202, USA
| |
Collapse
|
26
|
Liu C, Shen Y, Cavdar O, Huang J, Fang H. Angiotensin II-induced vascular endothelial cells ferroptosis via P53-ALOX12 signal axis. Clin Exp Hypertens 2023; 45:2180019. [PMID: 36860117 DOI: 10.1080/10641963.2023.2180019] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
OBJECTIVES The present study aimed to investigate the effect and mechanism of angiotensin II-induced ferroptosis in vascular endothelial cells. METHODS In vitro, HUVECs were treated with AngII, AT1/2 R antagonist, P53 inhibitor, or their combinations. MDA and intracellular iron content were evaluated using an ELISA assay. The expression of ALOX12, P53, P21, and SLC7A11 were determined by western blotting in HUVECs and then confirmed through RT-PCR. RESULTS As the concentration of Ang II (0, 0.1,1,10,100, and 1000uM for 48 h) increased, the level of MDA and intracellular iron content increased in HUVECs. Compared with the single AngII group, ALOX12, p53, MDA, and intracellular iron content in AT1/2R antagonist group decreased significantly. In pifithrin-α hydrobromide-treated, ALOX12, P21,MDA, and intracellular iron content decreased significantly as compared to the single AngII group. Similarly, the effect of combined use of blockers is stronger than that of blockers alone. CONCLUSIONS AngII can induce ferroptosis of vascular endothelial cells. The mechanism of AngII-induced ferroptosis may be regulated through the signal axis of p53-ALOX12.
Collapse
Affiliation(s)
- Chi Liu
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Jing an District Central Hospital, Shanghai, China
| | - Yi Shen
- Department of General Practice, Pudong New Area District Zhoupu Hospital, Shanghai, China
| | - Omer Cavdar
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junling Huang
- Department of Geriatrics, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hong Fang
- Department of Cardiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
27
|
Mohamed R, Sullivan JC. Sustained activation of 12/15 lipoxygenase (12/15 LOX) contributes to impaired renal recovery post ischemic injury in male SHR compared to females. Mol Med 2023; 29:163. [PMID: 38049738 PMCID: PMC10696802 DOI: 10.1186/s10020-023-00762-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/19/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI) due to ischemia-reperfusion (IR) is a serious and frequent complication in clinical settings, and mortality rates remain high. There are well established sex differences in renal IR, with males exhibiting greater injury following an ischemic insult compared to females. We recently reported that males have impaired renal recovery from ischemic injury vs. females. However, the mechanisms mediating sex differences in renal recovery from IR injury remain poorly understood. Elevated 12/15 lipoxygenase (LOX) activity has been reported to contribute to the progression of numerous kidney diseases. The goal of the current study was to test the hypothesis that enhanced activation of 12/15 LOX contributes to impaired recovery post-IR in males vs. females. METHODS 13-week-old male and female spontaneously hypertensive rats (SHR) were randomized to sham or 30-minute warm bilateral IR surgery. Additional male and female SHR were randomized to treatment with vehicle or the specific 12/15 LOX inhibitor ML355 1 h prior to sham/IR surgery, and every other day following up to 7-days post-IR. Blood was collected from all rats 1-and 7-days post-IR. Kidneys were harvested 7-days post-IR and processed for biochemical, histological, and Western blot analysis. 12/15 LOX metabolites 12 and 15 HETE were measured in kidney samples by liquid chromatography-mass spectrometry (LC/MS). RESULTS Male SHR exhibited delayed recovery of renal function post-IR vs. male sham and female IR rats. Delayed recovery in males was associated with activation of renal 12/15 LOX, increased renal 12-HETE, enhanced endoplasmic reticulum (ER) stress, lipid peroxidation, renal cell death and inflammation compared to females 7-days post-IR. Treatment of male SHR with ML355 lowered levels of 12-HETE and resulted in reduced renal lipid peroxidation, ER stress, tubular cell death and inflammation 7-days post-IR with enhanced recovery of renal function compared to vehicle-treated IR male rats. ML355 treatment did not alter IR-induced increases in plasma creatinine in females, however, tubular injury and cell death were attenuated in ML355 treated females compared to vehicle-treated rats 7 days post-IR. CONCLUSION Our data demonstrate that sustained activation 12/15 LOX contributes to impaired renal recovery post ischemic injury in male and female SHR, although males are more susceptible on this mechanism than females.
Collapse
Affiliation(s)
- Riyaz Mohamed
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, 30912, United States.
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, 30912, United States
| |
Collapse
|
28
|
Schartz D, Akkipeddi SMK, Chittaranjan S, Rahmani R, Gunturi A, Ellens N, Kohli GS, Kessler A, Mattingly T, Morrell C, Bhalla T, Bender MT. CT hyperdense cerebral artery sign reflects distinct proteomic composition in acute ischemic stroke thrombus. J Neurointerv Surg 2023; 15:1264-1268. [PMID: 36878687 DOI: 10.1136/jnis-2022-019937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/17/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND Hyperdense cerebral artery sign (HCAS) is an imaging biomarker in acute ischemic stroke (AIS) that has been shown to be associated with various clinical outcomes and stroke etiology. While prior studies have correlated HCAS with histopathological composition of cerebral thrombus, it is unknown whether and to what extent HCAS is also associated with distinct clot protein composition. METHODS Thromboembolic material from 24 patients with AIS were retrieved via mechanical thrombectomy and evaluated with mass spectrometry in order to characterize their proteomic composition. Presence (+) or absence (-) of HCAS on preintervention non-contrast head CT was then determined and correlated with thrombus protein signature with abundance of individual proteins calculated as a function HCAS status. RESULTS 24 clots with 1797 distinct proteins in total were identified. 14 patients were HCAS(+) and 10 were HCAS(-). HCAS(+) were most significantly differentially abundant in actin cytoskeletal protein (P=0.002, Z=2.82), bleomycin hydrolase (P=0.007, Z=2.44), arachidonate 12-lipoxygenase (P=0.004, Z=2.60), and lysophospholipase D (P=0.007, Z=2.44), among other proteins; HCAS(-) clots were differentially enriched in soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein (P=0.0009, Z=3.11), tyrosine-protein kinase Fyn (P=0.002, Z=2.84), and several complement proteins (P<0.05, Z>1.71 for all), among numerous other proteins. Additionally, HCAS(-) thrombi were enriched in biological processes involved with plasma lipoprotein and protein-lipid remodeling/assembling, and lipoprotein metabolic processes (P<0.001), as well as cellular components including mitochondria (P<0.001). CONCLUSIONS HCAS is reflective of distinct proteomic composition in AIS thrombus. These findings suggest that imaging can be used to identify mechanisms of clot formation or maintenance at the protein level, and might inform future research on thrombus biology and imaging characterization.
Collapse
Affiliation(s)
- Derrek Schartz
- Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Sajal Medha K Akkipeddi
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Siddharth Chittaranjan
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Redi Rahmani
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Aditya Gunturi
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Nathaniel Ellens
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Gurkirat Singh Kohli
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Alex Kessler
- Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Thomas Mattingly
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Craig Morrell
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Tarun Bhalla
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Matthew T Bender
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
29
|
Chen B, Zhao L, Yang R, Xu T. The recent advancements of ferroptosis in the diagnosis, treatment and prognosis of ovarian cancer. Front Genet 2023; 14:1275154. [PMID: 38028615 PMCID: PMC10665572 DOI: 10.3389/fgene.2023.1275154] [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: 08/09/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Ovarian cancer affects the female reproductive system and is the primary cause of cancer related mortality globally. The imprecise and non-specific nature of ovarian cancer symptoms often results in patients being diagnosed at an advanced stage, with metastatic lesions extending beyond the ovary. This presents a significant clinical challenge and imposes a substantial economic burden on both patients and society. Despite advancements in surgery, chemotherapy, and immunotherapy, the prognosis for most patients with ovarian cancer remains unsatisfactory. Therefore, the development of novel treatment strategies is imperative. Ferroptosis, a distinct form of regulated cell death, characterized by iron-dependent lipid peroxidation, differs from autophagy, apoptosis, and necrosis, and may hold promise as a novel cell death. Numerous studies have demonstrated the involvement of ferroptosis in various conventional signaling pathways and biological processes. Recent investigations have revealed the significant contribution of ferroptosis in the initiation, progression, and metastasis of diverse malignant tumors, including ovarian cancer. Moreover, ferroptosis exhibits a synergistic effect with chemotherapy, radiotherapy, and immunotherapy in restraining the proliferation of ovarian cancer cells. The aforementioned implies that ferroptosis holds considerable importance in the management of ovarian cancer and has the potential to serve as a novel therapeutic target. The present review provides a comprehensive overview of the salient features of ferroptosis, encompassing its underlying mechanisms and functional role in ovarian cancer, along with the associated signaling pathways and genes. Furthermore, the review highlights the prospective utility of ferroptosis in the treatment of ovarian cancer.
Collapse
Affiliation(s)
| | | | | | - Tianmin Xu
- The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Chi J, Cheng J, Wang S, Li C, Chen M. Promising Anti-Inflammatory Tools: Biomedical Efficacy of Lipoxins and Their Synthetic Pathways. Int J Mol Sci 2023; 24:13282. [PMID: 37686088 PMCID: PMC10487465 DOI: 10.3390/ijms241713282] [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: 06/27/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 09/10/2023] Open
Abstract
Lipoxins (LXs) have attracted widespread attention as a class of anti-inflammatory lipid mediators that are produced endogenously by the organism. LXs are arachidonic acid (ARA) derivatives that include four different structures: lipoxin A4 (LXA4), lipoxin B4 (LXB4), and the aspirin-induced differential isomers 15-epi-LXA4 and 15-epi-LXB4. Because of their unique biological activity of reducing inflammation in the body, LXs have great potential for neuroprotection, anti-inflammatory treatment of COVID-19, and other related diseases. The synthesis of LXs in vivo is achieved through the action of lipoxygenase (LO). As a kind of important enzyme, LO plays a major role in the physiological processes of living organisms in mammals and functions in some bacteria and fungi. This suggests new options for the synthesis of LXs in vitro. Meanwhile, there are other chemical and biochemical methods to synthesize LXs. In this review, the recent progress on physiological activity and synthetic pathways of LXs is summarized, and new insights into the synthesis of LXs in vitro are provided.
Collapse
Affiliation(s)
| | | | | | | | - Ming Chen
- School of Biological Engineering, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
31
|
Falsetta ML, Maddipati KR, Honn KV. Inflammation, lipids, and pain in vulvar disease. Pharmacol Ther 2023; 248:108467. [PMID: 37285943 PMCID: PMC10527276 DOI: 10.1016/j.pharmthera.2023.108467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
Localized provoked vulvodynia (LPV) affects ∼14 million people in the US (9% of women), destroying lives and relationships. LPV is characterized by chronic pain (>3 months) upon touch to the vulvar vestibule, which surrounds the vaginal opening. Many patients go months or years without a diagnosis. Once diagnosed, the treatments available only manage the symptoms of disease and do not correct the underlying problem. We have focused on elucidating the underlying mechanisms of chronic vulvar pain to speed diagnosis and improve intervention and management. We determined the inflammatory response to microorganisms, even members of the resident microflora, sets off a chain of events that culminates in chronic pain. This agrees with findings from several other groups, which show inflammation is altered in the painful vestibule. The vestibule of patients is acutely sensitive to inflammatory stimuli to the point of being deleterious. Rather than protect against vaginal infection, it causes heightened inflammation that does not resolve, which coincides with alterations in lipid metabolism that favor production of proinflammatory lipids and not pro-resolving lipids. Lipid dysbiosis in turn triggers pain signaling through the transient receptor potential vanilloid subtype 4 receptor (TRPV4). Treatment with specialized pro-resolving mediators (SPMs) that foster resolution reduces inflammation in fibroblasts and mice and vulvar sensitivity in mice. SPMs, specifically maresin 1, act on more than one part of the vulvodynia mechanism by limiting inflammation and acutely inhibiting TRPV4 signaling. Therefore, SPMs or other agents that target inflammation and/or TRPV4 signaling could prove effective as new vulvodynia therapies.
Collapse
Affiliation(s)
- Megan L Falsetta
- University of Rochester, OB/GYN Research Division, Rochester, NY, United States of America; University of Rochester, Pharmacology and Physiology Department, Rochester, NY, United States of America.
| | - Krishna Rao Maddipati
- Wayne State University, Pathology Department, Detroit, MI, United States of America; Wayne State University, Lipidomics Core Facility and Bioactive Lipids Research Program, Detroit, MI, United States of America
| | - Kenneth V Honn
- Wayne State University, Pathology Department, Detroit, MI, United States of America; Wayne State University, Lipidomics Core Facility and Bioactive Lipids Research Program, Detroit, MI, United States of America
| |
Collapse
|
32
|
Wang X, Liang T, Mao Y, Li Z, Li X, Zhu X, Cao F, Zhang J. Nervonic acid improves liver inflammation in a mouse model of Parkinson's disease by inhibiting proinflammatory signaling pathways and regulating metabolic pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154911. [PMID: 37276724 DOI: 10.1016/j.phymed.2023.154911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND Nervonic acid (NA) - a type of bioactive fatty acid that is found in natural sources - can inhibit inflammatory reactions and regulate immune system balance. Therefore, the use of NA for the treatment of neurodegenerative diseases has received considerable attention. Our previous study found that NA inhibited inflammatory responses in the brain of Parkinson's disease (PD) mouse models. In addition to the brain, PD is also associated with visceral organ dysfunction, especially impaired liver function. Thus, studying the role of NA in PD-mediated inflammation of the liver is particularly important. METHODS A combined transcriptome and metabolomic approach was utilized to investigate the anti-inflammatory effects of NA on the liver of PD mice. Inflammatory signaling molecules and metabolic pathway-related genes were examined in the liver using real-time PCR and western blotting. RESULTS Liver transcriptome analysis revealed that NA exerted anti-inflammatory effects by controlling several pro-inflammatory signaling pathways, such as the down-regulation of the tumor necrosis factor and nuclear factor kappa B signaling pathways, both of which were essential in the development of inflammatory disease. In addition, liver metabolomic results revealed that metabolites related to steroid hormone biosynthesis, arachidonic acid metabolism, and linoleic acid metabolism were up-regulated and those related to valine, leucine, and isoleucine degradation pathways were down-regulated in NA treatment groups compared with the PD model. The integration of metabolomic and transcriptomic results showed NA significantly exerted its anti-inflammatory function by regulating the transcription and metabolic pathways of multiple genes. Particularly, linoleic acid metabolism, arachidonic acid metabolism, and steroid hormone biosynthesis were the crucial pathways of the anti-inflammatory action of NA. Key genes in these metabolic pathways and key molecules in inflammatory signaling pathways were also verified, which were consistent with transcriptomic results. CONCLUSION These findings provide novel insights into the liver protective effects of NA against PD mice. This study also showed that NA could be a useful dietary element for improving and treating PD-induced liver inflammation.
Collapse
Affiliation(s)
- Xueqi Wang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Tingyu Liang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Ying Mao
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Zhengdou Li
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Xu Li
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Xinliang Zhu
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China; Bioactive Products Engineering Research Center for Gansu Distinctive Plants, Lanzhou 730070, China; Institute of Rural Development and Research, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Fuliang Cao
- Nanjing Forestry University, Nanjing, Jiangsu Province 210037, China.
| | - Ji Zhang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China; Bioactive Products Engineering Research Center for Gansu Distinctive Plants, Lanzhou 730070, China; Institute of Rural Development and Research, Northwest Normal University, Lanzhou, Gansu Province 730070, China.
| |
Collapse
|
33
|
Zhang T, Hu X, Yu S, Wei C. Construction of ceRNA network based on RNA-seq for identifying prognostic lncRNA biomarkers in Perthes disease. Front Genet 2023; 14:1105893. [PMID: 37303951 PMCID: PMC10252144 DOI: 10.3389/fgene.2023.1105893] [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: 11/23/2022] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction: Legg-Calvé-Perthes disease or Perthes disease is a condition that occurs in children aged 2 to 15 years, and is characterized by osteonecrosis of the femoral head, which results in physical limitations. Despite ongoing research, the pathogenesis and molecular mechanisms underlying the development of Perthes disease remain unclear. In order to obtain further insights, the expression patterns of long non-coding RNAs (lncRNAs), miRNAs, and mRNAs in a rabbit model of Perthes disease were analyzed in this study by transcriptome sequencing. Methods and results: The results of RNA-seq analyses revealed that 77 lncRNAs, 239 miRNAs, and 1027 mRNAs were differentially expressed in the rabbit model. This finding suggested that multiple genetic pathways are involved in the development of Perthes disease. A weighted gene co-expression network analysis (WGCNA) network was subsequently constructed using the differentially expressed mRNAs (DEmRNAs), and network analysis revealed that the genes associated with angiogenesis and platelet activation were downregulated, which was consistent with the findings of Perthes disease. A competing endogenous RNA (ceRNA) network was additionally constructed using 29 differentially expressed lncRNAs (including HIF3A and LOC103350994), 28 differentially expressed miRNAs (including ocu-miR-574-5p and ocu-miR-324-3p), and 76 DEmRNAs (including ALOX12 and PTGER2). Disscusion: The results obtained herein provide novel perspectives regarding the pathogenesis and molecular mechanisms underlying the development of Perthes disease. The findings of this study can pave the way for the development of effective therapeutic strategies for Perthes disease in future.
Collapse
Affiliation(s)
- Tianjiu Zhang
- Guizhou Children’s Hospital, Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiaolin Hu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Song Yu
- Guizhou Children’s Hospital, Department of Pediatric Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Chunyan Wei
- Department of Gynecoloay, Obstetrics and Gynecoloay Hospital of Fudan University, Shanchai, China
| |
Collapse
|
34
|
León-Reyes G, Argoty-Pantoja AD, Becerra-Cervera A, López-Montoya P, Rivera-Paredez B, Velázquez-Cruz R. Oxidative-Stress-Related Genes in Osteoporosis: A Systematic Review. Antioxidants (Basel) 2023; 12:antiox12040915. [PMID: 37107290 PMCID: PMC10135393 DOI: 10.3390/antiox12040915] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Osteoporosis is characterized by a decline in bone mineral density (BMD) and increased fracture risk. Free radicals and antioxidant systems play a central role in bone remodeling. This study was conducted to illustrate the role of oxidative-stress-related genes in BMD and osteoporosis. A systematic review was performed following the PRISMA guidelines. The search was computed in PubMed, Web of Sciences, Scopus, EBSCO, and BVS from inception to November 1st, 2022. The risk of bias was evaluated using the Joanna Briggs Institute Critical Appraisal Checklist tool. A total of 427 potentially eligible articles exploring this search question were detected. After removing duplicates (n = 112) and excluding irrelevant manuscripts based on screenings of their titles and abstracts (n = 317), 19 articles were selected for full-text review. Finally, 14 original articles were included in this systematic review after we applied the exclusion and inclusion criteria. Data analyzed in this systematic review indicated that oxidative-stress-related genetic polymorphisms are associated with BMD at different skeletal sites in diverse populations, influencing the risk of osteoporosis or osteoporotic fracture. However, it is necessary to look deep into their association with bone metabolism to determine if the findings can be translated into the clinical management of osteoporosis and its progression.
Collapse
Affiliation(s)
- Guadalupe León-Reyes
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico
| | - Anna D Argoty-Pantoja
- Research Center in Policies, Population and Health, School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | - Adriana Becerra-Cervera
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico
- National Council of Science and Technology (CONACYT), Mexico City 03940, Mexico
| | - Priscilla López-Montoya
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico
| | - Berenice Rivera-Paredez
- Research Center in Policies, Population and Health, School of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | - Rafael Velázquez-Cruz
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico
| |
Collapse
|
35
|
Ishimwe JA, Ferguson JF, Kirabo A. Sex Differences in Fatty Acid Metabolism and Blood Pressure Response to Dietary Salt in Humans. CARDIOGENETICS 2023; 13:33-46. [PMID: 38605973 PMCID: PMC11008634 DOI: 10.3390/cardiogenetics13010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024] Open
Abstract
Salt sensitivity is a trait in which high dietary sodium (Na+) intake causes an increase in blood pressure (BP). We previously demonstrated that in the gut, elevated dietary Na+ causes dysbiosis. The mechanistic interplay between excess dietary Na+-induced alteration in the gut microbiome and sex differences is less understood. The goal of this study was to identify novel metabolites in sex differences and blood pressure in response to a high dietary Na+ intake. We performed stool and plasma metabolomics analysis and measured the BP of human volunteers with salt intake above or below the American Heart Association recommendations. We also performed RNA sequencing on human monocytes treated with high salt in vitro. The relationship between BP and dietary Na+ intake was different in women and men. Network analysis revealed that fatty acids as top subnetworks differentially changed with salt intake. We found that women with high dietary Na+ intake have high levels of arachidonic acid related metabolism, suggesting a role in sex differences of the blood pressure response to Na+. The exposure of monocytes to high salt in vitro upregulates the transcription of fatty acid receptors and arachidonic acid-related genes. These findings provide potentially novel insights into metabolic changes underlying gut dysbiosis and inflammation in salt sensitivity of BP.
Collapse
Affiliation(s)
- Jeanne A. Ishimwe
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
| | - Jane F. Ferguson
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Nashville, TN 37235, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37235, USA
- Medical Center, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
36
|
Selective ischemic-hemisphere targeting Ginkgolide B liposomes with improved solubility and therapeutic efficacy for cerebral ischemia-reperfusion injury. Asian J Pharm Sci 2023; 18:100783. [PMID: 36891470 PMCID: PMC9986716 DOI: 10.1016/j.ajps.2023.100783] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/23/2022] [Accepted: 01/24/2023] [Indexed: 02/13/2023] Open
Abstract
Cerebral ischemia-reperfusion injury (CI/RI) remains the main cause of disability and death in stroke patients due to lack of effective therapeutic strategies. One of the main issues related to CI/RI treatment is the presence of the blood-brain barrier (BBB), which affects the intracerebral delivery of drugs. Ginkgolide B (GB), a major bioactive component in commercially available products of Ginkgo biloba, has been shown significance in CI/RI treatment by regulating inflammatory pathways, oxidative damage, and metabolic disturbance, and seems to be a candidate for stroke recovery. However, limited by its poor hydrophilicity and lipophilicity, the development of GB preparations with good solubility, stability, and the ability to cross the BBB remains a challenge. Herein, we propose a combinatorial strategy by conjugating GB with highly lipophilic docosahexaenoic acid (DHA) to obtain a covalent complex GB-DHA, which can not only enhance the pharmacological effect of GB, but can also be encapsulated in liposomes stably. The amount of finally constructed Lipo@GB-DHA targeting to ischemic hemisphere was validated 2.2 times that of free solution in middle cerebral artery occlusion (MCAO) rats. Compared to the marketed ginkgolide injection, Lipo@GB-DHA significantly reduced infarct volume with better neurobehavioral recovery in MCAO rats after being intravenously administered both at 2 h and 6 h post-reperfusion. Low levels of reactive oxygen species (ROS) and high neuron survival in vitro was maintained via Lipo@GB-DHA treatment, while microglia in the ischemic brain were polarized from the pro-inflammatory M1 phenotype to the tissue-repairing M2 phenotype, which modulate neuroinflammatory and angiogenesis. In addition, Lipo@GB-DHA inhibited neuronal apoptosis via regulating the apoptotic pathway and maintained homeostasis by activating the autophagy pathway. Thus, transforming GB into a lipophilic complex and loading it into liposomes provides a promising nanomedicine strategy with excellent CI/RI therapeutic efficacy and industrialization prospects.
Collapse
|
37
|
Cheng Z, Chen Y, Huang H. Identification and Validation of a Novel Prognostic Signature Based on Ferroptosis-Related Genes in Ovarian Cancer. Vaccines (Basel) 2023; 11:vaccines11020205. [PMID: 36851083 PMCID: PMC9962729 DOI: 10.3390/vaccines11020205] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Ovarian cancer is the most lethal gynecological tumor, with a poor prognosis due to the lack of early symptoms, resistance to chemotherapy, and recurrence. Ferroptosis belongs to the regulated cell death family, and is characterized by iron-dependent processes. Here, comprehensive bioinformatics analysis was applied to explore a valuable prognostic model based on ferroptosis-related genes, which was further validated in clinical OC samples. METHODS mRNA data of normal and ovarian tumor samples were obtained separately from the GTEx and TCGA databases. The least absolute shrinkage and selection operator (LASSO) cox regression was applied to construct the prognostic model based on ferroptosis-associated genes. Expression of ALOX12 in OC cell lines, as well as cell functions, including proliferation and migration, were examined. Finally, the prognostic efficiency of the model was assessed in the clinical tissues of OC patients. RESULTS A gene signature consisting of ALOX12, RB1, DNAJB6, STEAP3, and SELENOS was constructed. The signature divided TCGA, ICGC, and GEO cohorts into high-risk and low-risk groups separately. Receiver operating characteristic (ROC) curves and independent prognostic factor analysis were carried out, and the prognostic efficacy was validated. The expression levels of ALOX12 in cell lines were examined. Inhibition of ALOX12 attenuated cell proliferation and migration in HEY cells. Moreover, the prognostic value of ALOX12 expression was examined in clinical samples of OC patients. CONCLUSION This work constructed a novel ferroptosis-associated gene model. Furthermore, the clinical predictive role of ALOX12 was identified in OC patients, suggesting that ALOX12 might act as a potential prognostic tool and therapeutic target for OC patients.
Collapse
Affiliation(s)
- Zhe Cheng
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Huichao Huang
- Department of Infectious Disease, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Correspondence:
| |
Collapse
|
38
|
Tran M, Signorelli RL, Yamaguchi A, Chen E, Holinstat M, Iavarone AT, Offenbacher AR, Holman T. Biochemical and hydrogen-deuterium exchange studies of the single nucleotide polymorphism Y649C in human platelet 12-lipoxygenase linked to a bleeding disorder. Arch Biochem Biophys 2023; 733:109472. [PMID: 36442529 PMCID: PMC9888433 DOI: 10.1016/j.abb.2022.109472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Human platelet 12-lipoxygenase (h12-LOX) is responsible for the formation of oxylipin products that play an important role in platelet aggregation. Single nucleotide polymorphisms (SNPs) of h12-LOX have been implicated in several diseases. In this study, we investigate the structural, dynamical, and functional impact of a h12-LOX SNP that generates a tyrosine-to-cysteine mutation at a buried site (Y649C h12-LOX) and was previously ascribed with reduced levels of 12(S)-hydroxyeicosatetraenoic acid (12S-HETE) production in isolated platelets. Herein, in vitro Michaelis-Menten kinetics show reduced catalytic rates for Y649C compared to WT h12-LOX at physiological or lower temperatures. Both proteins exhibited similar melting temperatures, metal content, and oligomerization state. Liposome binding for both proteins was also dependent upon the presence of calcium, temperature, and liposome composition; however, the Y649C variant was found to have lowered binding capacity to liposomes compared to WT at physiological temperatures. Further, hydrogen-deuterium exchange mass spectrometry (HDX-MS) experiments revealed a regional defined enhancement in the peptide mobility caused by the mutation. This increased instability for the mutation stemmed from a change in an interaction with an arched helix that lines the substrate binding site, located ≥15 Å from the mutation site. Finally, differential scanning calorimetry demonstrated a reduced protein (un)folding enthalpy, consistent with the HDX results. Taken together, these results demonstrate remarkable similarity between the mutant and WT h12-LOX, and yet, subtle changes in activity, membrane affinity and protein stability may be responsible for the significant physiological changes that the Y649C SNP manifests in platelet biology.
Collapse
Affiliation(s)
- Michelle Tran
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, 95064, USA
| | | | - Adriana Yamaguchi
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Eefie Chen
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Anthony T. Iavarone
- QB3/Chemistry Mass Spectrometry Facility, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Adam R. Offenbacher
- Department of Chemistry, East Carolina University, Greenville, NC, 27858, USA,Corresponding author. (A.R. Offenbacher)
| | - Theodore Holman
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, 95064, USA,Corresponding author. (T. Holman)
| |
Collapse
|
39
|
Pablo-Torres C, Izquierdo E, Tan TJ, Obeso D, Layhadi JA, Sánchez-Solares J, Mera-Berriatua L, Bueno-Cabrera JL, Del Mar Reaño-Martos M, Iglesias-Cadarso A, Barbas C, Gomez-Casado C, Villaseñor A, Barber D, Shamji MH, Escribese MM. Deciphering the role of platelets in severe allergy by an integrative omics approach. Allergy 2022; 78:1319-1332. [PMID: 36527294 DOI: 10.1111/all.15621] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/04/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Mechanisms causing the onset and perpetuation of inflammation in severe allergic patients remain unknown. Our previous studies suggested that severe allergic inflammation is linked to platelet dysfunction. METHODS Platelet-rich plasma (PRP) and platelet-poor plasma (PPP) samples were obtained by platelet-apheresis from severe (n = 7) and mild (n = 10) allergic patients and nonallergic subjects (n = 9) to perform platelet lipidomics by liquid chromatography coupled to mass spectrometry (LC-MS) and RNA-seq analysis. Significant metabolites and transcripts were used to identify compromised biological pathways in the severe phenotype. Platelet and inflammation-related proteins were quantified by Luminex. RESULTS Platelets from severe allergic patients were characterized by high levels of ceramides, phosphoinositols, phosphocholines, and sphingomyelins. In contrast, they showed a decrease in eicosanoid precursor levels. Biological pathway analysis performed with the significant lipids revealed the alteration of phospholipases, calcium-dependent events, and linolenic metabolism. RNAseq confirmed mRNA overexpression of genes related to platelet activation and arachidonic acid metabolism in the severe phenotypes. Pathway analysis indicated the alteration of NOD, MAPK, TLR, TNF, and IL-17 pathways in the severe phenotype. P-Selectin and IL-17AF proteins were increased in the severe phenotype. CONCLUSIONS This study demonstrates that platelet lipid, mRNA, and protein content is different according to allergy severity. These findings suggest that platelet load is a potential source of biomarkers and a new chance for therapeutic targets in severe inflammatory pathologies.
Collapse
Affiliation(s)
- Carmela Pablo-Torres
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España
| | - Elena Izquierdo
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España
| | - Tiak Ju Tan
- National Heart and Lung Institute, Allergy and Clinical Immunology, Imperial College NIHR Biomedical Research Centre, Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - David Obeso
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España.,Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España
| | - Janice A Layhadi
- National Heart and Lung Institute, Allergy and Clinical Immunology, Imperial College NIHR Biomedical Research Centre, Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Javier Sánchez-Solares
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España
| | - Leticia Mera-Berriatua
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España
| | - José Luis Bueno-Cabrera
- Department of Hematology and Hemotherapy, Puerta de Hierro-Majadahonda University Hospital, Madrid, Spain
| | | | - Alfredo Iglesias-Cadarso
- Department of Allergy and Immunology, Puerta de Hierro-Majadahonda University Hospital, Madrid, Spain
| | - Coral Barbas
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España
| | - Cristina Gomez-Casado
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España
| | - Alma Villaseñor
- Centro de Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España
| | - Domingo Barber
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España
| | - Mohamed H Shamji
- National Heart and Lung Institute, Allergy and Clinical Immunology, Imperial College NIHR Biomedical Research Centre, Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - María M Escribese
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Boadilla del Monte, España
| |
Collapse
|
40
|
Lee TJ, Nettleford SK, McGlynn A, Carlson BA, Kirimanjeswara GS, Prabhu KS. The role of selenoproteins in neutrophils during inflammation. Arch Biochem Biophys 2022; 732:109452. [PMID: 36336122 PMCID: PMC9712253 DOI: 10.1016/j.abb.2022.109452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/22/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Polymorphonuclear neutrophils (PMNs)-derived ROS are involved in the regulation of multiple functions of PMNs critical in both inflammation and its timely resolution. Selenium is an essential trace element that functions as a gatekeeper of cellular redox homeostasis in the form of selenoproteins. Despite their well-studied involvement in regulating functions of various immune cells, limited studies have focused on the regulation of selenoproteins in PMN and their associated functions. Ex-vivo treatment of murine primary bone marrow derived PMNs with bacterial endotoxin lipopolysaccharide (LPS) indicated temporal regulation of several selenoprotein genes at the mRNA level. However, only glutathione peroxidase 4 (Gpx4) was significantly upregulated, while Selenof, Selenow, and Gpx1 were significantly downregulated in a temporal manner at the protein level. Exposure of PMNs isolated from tRNASec (Trsp)fl/fl S100A8Cre (TrspN) PMN-specific selenoprotein knockout mice, to the Gram-negative bacterium, Citrobacter rodentium, showed decreased bacterial growth, reduced phagocytosis, as well as impaired neutrophil extracellular trap (NET) formation ability, when compared to the wild-type PMNs. Increased extracellular ROS production upon LPS stimulation was also observed in TrspN PMNs that was associated with upregulation of Alox12, Cox2, and iNOS, as well as proinflammatory cytokines such as TNFα and IL-1β. Our data indicate that the inhibition of selenoproteome expression results in alteration of PMN proinflammatory functions, suggesting a potential role of selenoproteins in the continuum of inflammation and resolution.
Collapse
Affiliation(s)
- Tai-Jung Lee
- Department of Veterinary & Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Shaneice K Nettleford
- Department of Veterinary & Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Allison McGlynn
- Department of Veterinary & Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Bradley A Carlson
- Office of Research Support, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Girish S Kirimanjeswara
- Department of Veterinary & Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, University Park, PA, 16802, USA
| | - K Sandeep Prabhu
- Department of Veterinary & Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
41
|
Wang X, Chen Y, Yang X, Cheng L, He Z, Xin Y, Huang S, Meng F, Zhang P, Luo L. Activation of ALOX12 by a multi-organelle-orienting photosensitizer drives ACSL4-independent cell ferroptosis. Cell Death Dis 2022; 13:1040. [PMID: 36517470 PMCID: PMC9751149 DOI: 10.1038/s41419-022-05462-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022]
Abstract
Ferroptosis is a recently-defined tumor suppression mechanism, but the sensitivity of many tumorigenic cells to ferroptosis is limited by their deficient expression of acyl-CoA synthetase long-chain family member 4 (ACSL4). Here, we report the discovery of a photosensitizer, namely TPCI, which can evoke ACSL4-independent ferroptosis of cancer cells in photodynamic therapy. Through co-localization with 12-lipoxygenase (ALOX12) in multiple subcellular organelles, TPCI activates ALOX12 to generate lipid reactive oxygen species in large quantity and trigger cell ferroptosis. Intriguingly, confining TPCI exclusively in lysosomes switches the cell death from ferroptosis to apoptosis. More strikingly, the ferroptosis mediated by TPCI-induced ALOX12 activation does not require the participation of ACSL4. Therefore, our study identifies TPCI as the first ALOX12 activator to induce ferroptosis independent of ACSL4, which renders a viable therapeutic approach on the basis of distinct ferroptosis of cancer cells, regardless their ACSL4 expressions.
Collapse
Affiliation(s)
- Xiuxia Wang
- grid.207374.50000 0001 2189 3846Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450052 China
| | - Yuanhong Chen
- grid.33199.310000 0004 0368 7223National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China
| | - Xiang Yang
- grid.33199.310000 0004 0368 7223National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China
| | - Lianghui Cheng
- grid.33199.310000 0004 0368 7223National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China
| | - Zhenyan He
- grid.33199.310000 0004 0368 7223National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China
| | - Yanru Xin
- grid.33199.310000 0004 0368 7223National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China
| | - Shan Huang
- grid.33199.310000 0004 0368 7223National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China
| | - Fanling Meng
- grid.33199.310000 0004 0368 7223National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China
| | - Peijing Zhang
- grid.33199.310000 0004 0368 7223National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China ,grid.33199.310000 0004 0368 7223Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China
| | - Liang Luo
- grid.33199.310000 0004 0368 7223National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China ,grid.33199.310000 0004 0368 7223Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074 China ,grid.33199.310000 0004 0368 7223Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074 China
| |
Collapse
|
42
|
Bacterial lipoxygenases: Biochemical characteristics, molecular structure and potential applications. Biotechnol Adv 2022; 61:108046. [DOI: 10.1016/j.biotechadv.2022.108046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/02/2022] [Accepted: 09/28/2022] [Indexed: 11/24/2022]
|
43
|
Chandrasekaran P, Negretti NM, Sivakumar A, Liberti DC, Wen H, Peers de Nieuwburgh M, Wang JY, Michki NS, Chaudhry FN, Kaur S, Lu M, Jin A, Zepp JA, Young LR, Sucre JMS, Frank DB. CXCL12 defines lung endothelial heterogeneity and promotes distal vascular growth. Development 2022; 149:dev200909. [PMID: 36239312 PMCID: PMC9687018 DOI: 10.1242/dev.200909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/22/2022] [Indexed: 11/05/2022]
Abstract
There is a growing amount of data uncovering the cellular diversity of the pulmonary circulation and mechanisms governing vascular repair after injury. However, the molecular and cellular mechanisms contributing to the morphogenesis and growth of the pulmonary vasculature during embryonic development are less clear. Importantly, deficits in vascular development lead to significant pediatric lung diseases, indicating a need to uncover fetal programs promoting vascular growth. To address this, we used a transgenic mouse reporter for expression of Cxcl12, an arterial endothelial hallmark gene, and performed single-cell RNA sequencing on isolated Cxcl12-DsRed+ endothelium to assess cellular heterogeneity within pulmonary endothelium. Combining cell annotation with gene ontology and histological analysis allowed us to segregate the developing artery endothelium into functionally and spatially distinct subpopulations. Expression of Cxcl12 is highest in the distal arterial endothelial subpopulation, a compartment enriched in genes for vascular development. Accordingly, disruption of CXCL12 signaling led to, not only abnormal branching, but also distal vascular hypoplasia. These data provide evidence for arterial endothelial functional heterogeneity and reveal conserved signaling mechanisms essential for pulmonary vascular development.
Collapse
Affiliation(s)
- Prashant Chandrasekaran
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Nicholas M. Negretti
- Department of Pediatrics, Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Aravind Sivakumar
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Derek C. Liberti
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Hongbo Wen
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Maureen Peers de Nieuwburgh
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Joanna Y. Wang
- Department of Medicine, University of Pennsylvania, Penn-CHOP Lung Biology Institute, Philadelphia, PA 19104, USA
| | - Nigel S. Michki
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Fatima N. Chaudhry
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Philadelphia, PA 19104, USA
| | - Sukhmani Kaur
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - MinQi Lu
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Annabelle Jin
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Philadelphia, PA 19104, USA
| | - Jarod A. Zepp
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Philadelphia, PA 19104, USA
| | - Lisa R. Young
- Department of Pediatrics, Division of Pulmonary and Sleep Medicine, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Philadelphia, PA 19104, USA
| | - Jennifer M. S. Sucre
- Department of Pediatrics, Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - David B. Frank
- Department of Pediatrics, Division of Cardiology, University of Pennsylvania, Children's Hospital of Philadelphia, Penn-CHOP Lung Biology Institute, Penn Cardiovascular Institute, Philadelphia, PA 19104, USA
| |
Collapse
|
44
|
Kotlyarov S. Genetic and Epigenetic Regulation of Lipoxygenase Pathways and Reverse Cholesterol Transport in Atherogenesis. Genes (Basel) 2022; 13:1474. [PMID: 36011386 PMCID: PMC9408222 DOI: 10.3390/genes13081474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is one of the most important medical and social problems of modern society. Atherosclerosis causes a large number of hospitalizations, disability, and mortality. A considerable amount of evidence suggests that inflammation is one of the key links in the pathogenesis of atherosclerosis. Inflammation in the vascular wall has extensive cross-linkages with lipid metabolism, and lipid mediators act as a central link in the regulation of inflammation in the vascular wall. Data on the role of genetics and epigenetic factors in the development of atherosclerosis are of great interest. A growing body of evidence is strengthening the understanding of the significance of gene polymorphism, as well as gene expression dysregulation involved in cross-links between lipid metabolism and the innate immune system. A better understanding of the genetic basis and molecular mechanisms of disease pathogenesis is an important step towards solving the problems of its early diagnosis and treatment.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
45
|
Weng S, Liu Z, Xu H, Ge X, Ren Y, Dang Q, Liu L, Zhang J, Luo P, Ren J, Han X. ALOX12: A Novel Insight in Bevacizumab Response, Immunotherapy Effect, and Prognosis of Colorectal Cancer. Front Immunol 2022; 13:910582. [PMID: 35833141 PMCID: PMC9271859 DOI: 10.3389/fimmu.2022.910582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer is a highly malignant cancer with poor prognosis and mortality rates. As the first biological agent approved for metastatic colorectal cancer (mCRC), bevacizumab was confirmed to exhibit good performance when combined with chemotherapy and immunotherapy. However, the efficacy of both bevacizumab and immunotherapy is highly heterogeneous across CRC patients with different stages. Thus, exploring a novel biomarker to comprehensively assess the prognosis and bevacizumab and immunotherapy response of CRC is of great significance. In our study, weighted gene co-expression network analysis (WGCNA) and the receiver operating characteristic (ROC) curves were employed to identify bevacizumab-related genes. After verification in four public cohorts and our internal cohort, ALOX12 was identified as a key gene related to bevacizumab response. Prognostic analysis and in vitro experiments further demonstrated that ALOX12 was closely associated with the prognosis, tumor proliferation, invasion, and metastasis. Multi-omics data analysis based on mutation and copy number variation (CNV) revealed that RYR3 drove the expression of ALOX12 and the deletion of 17p12 inhibited ALOX12 expression, respectively. Moreover, we interrogated the relationship between ALOX12 and immune cells and checkpoints. The results exhibited that high ALOX12 expression predicted a higher immune infiltration and better immunotherapy response, which was further validated in Tumor Immune Dysfunction and Exclusion (TIDE) and Subclass Mapping (SubMap) methods. Above all, our study provides a stable biomarker for clinical protocol optimization, prognostic assessment, precise treatment, and individualized treatment of CRC.
Collapse
Affiliation(s)
- Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
- *Correspondence: Xinwei Han, ; Jianzhuang Ren, ; Zaoqu Liu,
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
| | - Xiaoyong Ge
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qin Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianzhuang Ren
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Xinwei Han, ; Jianzhuang Ren, ; Zaoqu Liu,
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Interventional Institute of Zhengzhou University, Zhengzhou, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, China
- *Correspondence: Xinwei Han, ; Jianzhuang Ren, ; Zaoqu Liu,
| |
Collapse
|
46
|
Khirfan F, Jarrar Y, Al-Qirim T, Goh KW, Jarrar Q, Ardianto C, Awad M, Al-Ameer HJ, Al-Awaida W, Moshawih S, Ming LC. Analgesics Induce Alterations in the Expression of SARS-CoV-2 Entry and Arachidonic-Acid-Metabolizing Genes in the Mouse Lungs. Pharmaceuticals (Basel) 2022; 15:696. [PMID: 35745615 PMCID: PMC9227818 DOI: 10.3390/ph15060696] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/22/2022] [Accepted: 05/28/2022] [Indexed: 11/16/2022] Open
Abstract
Paracetamol and nonsteroidal anti-inflammatory drugs are widely used in the management of respiratory viral infections. This study aimed to determine the effects of the most commonly used analgesics (paracetamol, ibuprofen, and diclofenac) on the mRNA expression of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) entry and arachidonic-acid-metabolizing genes in mouse lungs. A total of twenty eight Balb/c mice were divided into four groups and treated separately with vehicle, paracetamol, ibuprofen, and diclofenac in clinically equivalent doses for 14 days. Then, the expressions of SARS-CoV-2 entry, ACE2, TMPRSS2, and Ctsl genes, in addition to the arachidonic-acid-metabolizing cyp450, cox, and alox genes, were analyzed using real-time PCR. Paracetamol increased the expressions of TMPRSS2 and Ctsl genes by 8.5 and 5.6 folds, respectively, while ibuprofen and diclofenac significantly decreased the expression of the ACE2 gene by more than 2.5 folds. In addition, all tested drugs downregulated (p < 0.05) cox2 gene expression, and paracetamol reduced the mRNA levels of cyp4a12 and 2j5. These molecular alterations in diclofenac and ibuprofen were associated with pathohistological alterations, where both analgesics induced the infiltration of inflammatory cells and airway wall thickening. It is concluded that analgesics such as paracetamol, ibuprofen, and diclofenac alter the expression of SARS-CoV-2 entry and arachidonic-acid-metabolizing genes in mouse lungs.
Collapse
Affiliation(s)
- Fatima Khirfan
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11731, Jordan; (F.K.); (T.A.-Q.); (M.A.)
| | - Yazun Jarrar
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11731, Jordan; (F.K.); (T.A.-Q.); (M.A.)
| | - Tariq Al-Qirim
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11731, Jordan; (F.K.); (T.A.-Q.); (M.A.)
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia;
| | - Qais Jarrar
- Department of Applied Pharmaceutical Sciences, Faculty of Pharmacy, Al-Isra University, Amman 11622, Jordan;
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Mohammad Awad
- Department of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11731, Jordan; (F.K.); (T.A.-Q.); (M.A.)
| | - Hamzeh J. Al-Ameer
- Department of Biology and Biotechnology, American University of Madaba, Madaba 17110, Jordan; (H.J.A.-A.); (W.A.-A.)
| | - Wajdy Al-Awaida
- Department of Biology and Biotechnology, American University of Madaba, Madaba 17110, Jordan; (H.J.A.-A.); (W.A.-A.)
| | - Said Moshawih
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei Darussalam;
| | - Long Chiau Ming
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
- PAP Rashidah Sa’adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei Darussalam;
| |
Collapse
|
47
|
Host cell proteins modulated upon Toxoplasma infection identified using proteomic approaches: a molecular rationale. Parasitol Res 2022; 121:1853-1865. [PMID: 35552534 DOI: 10.1007/s00436-022-07541-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
Toxoplasma gondii is a pathogenic protozoan parasite belonging to the apicomplexan phylum that infects the nucleated cells of warm-blooded hosts leading to an infectious disease known as toxoplasmosis. Apicomplexan parasites such as T. gondii can display different mechanisms to control or manipulate host cells signaling at different levels altering the host subcellular genome and proteome. Indeed, Toxoplasma is able to modulate host cell responses (especially immune responses) during infection to its advantage through both structural and functional changes in the proteome of different infected cells. Consequently, parasites can transform the invaded cells into a suitable environment for its own replication and the induction of infection. Proteomics as an applicable tool can identify such critical proteins involved in pathogen (Toxoplasma)-host cell interactions and consequently clarify the cellular mechanisms that facilitate the entry of pathogens into host cells, and their replication and transmission, as well as the central mechanisms of host defense against pathogens. Accordingly, the current paper reviews several proteins (identified using proteomic approaches) differentially expressed in the proteome of Toxoplasma-infected host cells (macrophages and human foreskin fibroblasts) and tissues (brain and liver) and highlights their plausible functions in the cellular biology of the infected cells. The identification of such modulated proteins and their related cell impact (cell responses/signaling) can provide further information regarding parasite pathogenesis and biology that might lead to a better understanding of therapeutic strategies and novel drug targets.
Collapse
|
48
|
Shpakova V, Rukoyatkina N, Al Arawe N, Prilepskaya A, Kharazova A, Sharina I, Gambaryan S, Martin E. ML355 Modulates Platelet Activation and Prevents ABT-737 Induced Apoptosis in Platelets. J Pharmacol Exp Ther 2022; 381:164-175. [PMID: 35197320 PMCID: PMC9073945 DOI: 10.1124/jpet.121.000973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/05/2022] [Indexed: 01/14/2023] Open
Abstract
12-lipoxigenase (12-LOX) is implicated in regulation of platelet activation processes and can be a new promising target for antiplatelet therapy. However, investigations of 12-LOX were restricted by the lack of specific and potent 12-LOX inhibitors and by controversial data concerning the role of 12-LOX metabolites in platelet functions. A novel specific 12-LOX inhibitor ML355 was shown to inhibit platelet aggregation without adverse side effects on hemostasis; however, the molecular mechanisms of its action on platelets are poorly understood. Here, we showed that ML355 inhibited platelet activation induced by thrombin or thromboxane A2, but not by collagen-related peptide. ML355 blocked protein kinase B, phosphoinositide 3-kinase, and extracellular signal-regulated kinase, but not p38 kinase, spleen tyrosine kinase (Syk), or phospholipase Cγ2 phosphorylation in activated platelets. The main inhibitory effect of low doses of ML355 (1-20 μM) on thrombin activated platelets was mediated by the decrease in reactive oxygen species level, whereas high doses of ML355 (50 μM) caused cyclic adenosine monophosphate activation. ML355 did not affect the activity of nitric oxide-dependent soluble guanylyl cyclase, nor did it affect the relaxation of preconstricted aortic rings in mice. ML355 itself did not affect platelet viability, but at 50 μM dose blocked caspase-dependent apoptosis induced by B-cell lymphoma II inhibitor ABT-737. SIGNIFICANCE STATEMENT: The current paper provides novel and original data concerning molecular mechanisms of 12-LOX inhibitor ML355 action on platelets. These data reveal antiplatelet and protective effects of ML355 on platelets and may be of importance for both antiplatelet and anticancer therapy.
Collapse
Affiliation(s)
- Valentina Shpakova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia (V.S., N.R., S.G.); Saint Petersburg State University, Saint Petersburg, Russia (N.A.A., A.P., A.K.); and Department of Internal Medicine, Division of Cardiology, University of Texas Houston Medical School, Houston, Texas (I.S., E.M.)
| | - Natalia Rukoyatkina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia (V.S., N.R., S.G.); Saint Petersburg State University, Saint Petersburg, Russia (N.A.A., A.P., A.K.); and Department of Internal Medicine, Division of Cardiology, University of Texas Houston Medical School, Houston, Texas (I.S., E.M.)
| | - Nada Al Arawe
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia (V.S., N.R., S.G.); Saint Petersburg State University, Saint Petersburg, Russia (N.A.A., A.P., A.K.); and Department of Internal Medicine, Division of Cardiology, University of Texas Houston Medical School, Houston, Texas (I.S., E.M.)
| | - Anna Prilepskaya
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia (V.S., N.R., S.G.); Saint Petersburg State University, Saint Petersburg, Russia (N.A.A., A.P., A.K.); and Department of Internal Medicine, Division of Cardiology, University of Texas Houston Medical School, Houston, Texas (I.S., E.M.)
| | - Alexandra Kharazova
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia (V.S., N.R., S.G.); Saint Petersburg State University, Saint Petersburg, Russia (N.A.A., A.P., A.K.); and Department of Internal Medicine, Division of Cardiology, University of Texas Houston Medical School, Houston, Texas (I.S., E.M.)
| | - Iraida Sharina
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia (V.S., N.R., S.G.); Saint Petersburg State University, Saint Petersburg, Russia (N.A.A., A.P., A.K.); and Department of Internal Medicine, Division of Cardiology, University of Texas Houston Medical School, Houston, Texas (I.S., E.M.)
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia (V.S., N.R., S.G.); Saint Petersburg State University, Saint Petersburg, Russia (N.A.A., A.P., A.K.); and Department of Internal Medicine, Division of Cardiology, University of Texas Houston Medical School, Houston, Texas (I.S., E.M.)
| | - Emil Martin
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg, Russia (V.S., N.R., S.G.); Saint Petersburg State University, Saint Petersburg, Russia (N.A.A., A.P., A.K.); and Department of Internal Medicine, Division of Cardiology, University of Texas Houston Medical School, Houston, Texas (I.S., E.M.)
| |
Collapse
|
49
|
Teh BP, Ahmad NB, Mohamad SB, Tan TYC, Mohd Abd Razak MRB, Afzan AB, Syed Mohamed AFB. Carica papaya Leaf Juice for Dengue: A Scoping Review. Nutrients 2022; 14:1584. [PMID: 35458146 PMCID: PMC9030784 DOI: 10.3390/nu14081584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
The potential therapeutic effect of Carica papaya leaf juice has attracted wide interest from the public and scientists in relieving dengue related manifestations. Currently, there is a lack of evaluated evidence on its juice form. Therefore, this scoping review aims to critically appraise the available scientific evidence related to the efficacy of C. papaya leaf juice in dengue. A systematic search was performed using predetermined keywords on two electronic databases (PubMed and Google Scholar). Searched results were identified, screened and appraised to establish the association between C. papaya and alleviating dengue associated conditions. A total of 28 articles (ethnobotanical information: three, in vitro studies: three, ex vivo studies: one, in vivo study: 13, clinical studies: 10) were included for descriptive analysis, which covered study characteristics, juice preparation/formulations, study outcomes, and toxicity findings. Other than larvicidal activity, this review also reveals two medicinal potentials of C. papaya leaf juice on dengue infection, namely anti-thrombocytopenic and immunomodulatory effects. C. papaya leaf juice has the potential to be a new drug candidate against dengue disease safely and effectively.
Collapse
Affiliation(s)
- Bee Ping Teh
- Herbal Medicine Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam 40170, Malaysia; (N.B.A.); (T.Y.C.T.); (M.R.B.M.A.R.); (A.B.A.); (A.F.B.S.M.)
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Norzahirah Binti Ahmad
- Herbal Medicine Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam 40170, Malaysia; (N.B.A.); (T.Y.C.T.); (M.R.B.M.A.R.); (A.B.A.); (A.F.B.S.M.)
| | - Saharuddin Bin Mohamad
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Centre of Research in Systems Biology, Structural Bioinformatics and Human Digital Imaging (CRYSTAL), Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Terence Yew Chin Tan
- Herbal Medicine Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam 40170, Malaysia; (N.B.A.); (T.Y.C.T.); (M.R.B.M.A.R.); (A.B.A.); (A.F.B.S.M.)
| | - Mohd Ridzuan Bin Mohd Abd Razak
- Herbal Medicine Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam 40170, Malaysia; (N.B.A.); (T.Y.C.T.); (M.R.B.M.A.R.); (A.B.A.); (A.F.B.S.M.)
| | - Adlin Binti Afzan
- Herbal Medicine Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam 40170, Malaysia; (N.B.A.); (T.Y.C.T.); (M.R.B.M.A.R.); (A.B.A.); (A.F.B.S.M.)
| | - Ami Fazlin Binti Syed Mohamed
- Herbal Medicine Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Shah Alam 40170, Malaysia; (N.B.A.); (T.Y.C.T.); (M.R.B.M.A.R.); (A.B.A.); (A.F.B.S.M.)
| |
Collapse
|
50
|
Sharma I, Liao Y, Zheng X, Kanwar YS. Modulation of gentamicin-induced acute kidney injury by myo-inositol oxygenase via the ROS/ALOX-12/12-HETE/GPR31 signaling pathway. JCI Insight 2022; 7:155487. [PMID: 35315361 PMCID: PMC8986073 DOI: 10.1172/jci.insight.155487] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 02/09/2022] [Indexed: 12/21/2022] Open
Abstract
In this investigation, a potentially novel signaling pathway in gentamicin-induced acute kidney injury-worsened by overexpression of proximal tubular enzyme, myo-inositol oxygenase (MIOX)-was elucidated. WT, MIOX-transgenic (MIOX-Tg), and MIOX-KO mice were used. Gentamicin was administered to induce tubular injury. MIOX-Tg mice had severe tubular lesions associated with increased serum creatinine and proteinuria. Lesions were relatively mild, with no rise in serum creatinine and no albuminuria in MIOX-KO mice. Transfection of HK-2 cells with MIOX-pcDNA led to increased gentamicin-induced reactive oxygen species (ROS). Marked increase of ROS-mediated lipid hydroperoxidation was noted in MIOX-Tg mice, as assessed by 4-HNE staining. This was associated with increased expression of arachidonate 12-lipoxygenase (ALOX-12) and generation of 12-hydroxyeicosatetraenoic acid (12-HETE). In addition, notable monocyte/macrophage influx, upregulation of NF-κB and inflammatory cytokines, and apoptosis was observed in MIOX-Tg mice. Treatment of cells with ALOX-12 siRNA abolished gentamicin-mediated induction of cytokines and 12-HETE generation. HETE-12 treatment promoted this effect, along with upregulation of various signaling kinases and activation of GPCR31. Similarly, treatment of cells or mice with the ALOX-12 inhibitor ML355 attenuated inflammatory response, kinase signaling cascade, and albuminuria. Collectively, these studies highlight a potentially novel mechanism (i.e., the ROS/ALOX-12/12-HETE/GPR31 signaling axis) relevant to gentamicin-induced nephrotoxicity modulated by MIOX.
Collapse
|