1
|
Jamil M, Cowart LA. Sphingolipids in mitochondria-from function to disease. Front Cell Dev Biol 2023; 11:1302472. [PMID: 38078003 PMCID: PMC10702779 DOI: 10.3389/fcell.2023.1302472] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/03/2023] [Indexed: 02/12/2024] Open
Abstract
Sphingolipids are not only structural components of cellular membranes but also play vital roles in cell signaling and modulation of cellular processes. Within mitochondria, sphingolipids exert diverse effects on mitochondrial dynamics, energy metabolism, oxidative stress, and cell death pathways. In this review, we summarize literature addressing the crucial role of sphingolipids in mitochondria, highlighting their impact on mitochondrial dynamics, cellular bioenergetics, and important cell processes including apoptosis and mitophagy.
Collapse
Affiliation(s)
- Maryam Jamil
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Lauren Ashley Cowart
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Richmond Veteran’s Affairs Medical Center, Richmond, VA, United States
| |
Collapse
|
2
|
Navarro-Lérida I, Aragay AM, Asensio A, Ribas C. Gq Signaling in Autophagy Control: Between Chemical and Mechanical Cues. Antioxidants (Basel) 2022; 11:1599. [PMID: 36009317 PMCID: PMC9405508 DOI: 10.3390/antiox11081599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
All processes in human physiology relies on homeostatic mechanisms which require the activation of specific control circuits to adapt the changes imposed by external stimuli. One of the critical modulators of homeostatic balance is autophagy, a catabolic process that is responsible of the destruction of long-lived proteins and organelles through a lysosome degradative pathway. Identification of the mechanism underlying autophagic flux is considered of great importance as both protective and detrimental functions are linked with deregulated autophagy. At the mechanistic and regulatory levels, autophagy is activated in response to diverse stress conditions (food deprivation, hyperthermia and hypoxia), even a novel perspective highlight the potential role of physical forces in autophagy modulation. To understand the crosstalk between all these controlling mechanisms could give us new clues about the specific contribution of autophagy in a wide range of diseases including vascular disorders, inflammation and cancer. Of note, any homeostatic control critically depends in at least two additional and poorly studied interdependent components: a receptor and its downstream effectors. Addressing the selective receptors involved in autophagy regulation is an open question and represents a new area of research in this field. G-protein coupled receptors (GPCRs) represent one of the largest and druggable targets membrane receptor protein superfamily. By exerting their action through G proteins, GPCRs play fundamental roles in the control of cellular homeostasis. Novel studies have shown Gαq, a subunit of heterotrimeric G proteins, as a core modulator of mTORC1 and autophagy, suggesting a fundamental contribution of Gαq-coupled GPCRs mechanisms in the control of this homeostatic feedback loop. To address how GPCR-G proteins machinery integrates the response to different stresses including oxidative conditions and mechanical stimuli, could provide deeper insight into new signaling pathways and open potential and novel therapeutic strategies in the modulation of different pathological conditions.
Collapse
Affiliation(s)
- Inmaculada Navarro-Lérida
- Molecular Biology Department and Center of Molecular Biology “Severo Ochoa”, CSIC-UAM, 28049 Madrid, Spain
- Health Research Institute “La Princesa”, 28006 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), ISCIII, 28029 Madrid, Spain
- Connexion Cancer-CSIC, 28006 Madrid, Spain
| | - Anna M. Aragay
- Department of Biology, Molecular Biology Institute of Barcelona (IBMB), Spanish National Research Council (CSIC), 08028 Barcelona, Spain
| | - Alejandro Asensio
- Molecular Biology Department and Center of Molecular Biology “Severo Ochoa”, CSIC-UAM, 28049 Madrid, Spain
- Health Research Institute “La Princesa”, 28006 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), ISCIII, 28029 Madrid, Spain
- Connexion Cancer-CSIC, 28006 Madrid, Spain
| | - Catalina Ribas
- Molecular Biology Department and Center of Molecular Biology “Severo Ochoa”, CSIC-UAM, 28049 Madrid, Spain
- Health Research Institute “La Princesa”, 28006 Madrid, Spain
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), ISCIII, 28029 Madrid, Spain
- Connexion Cancer-CSIC, 28006 Madrid, Spain
| |
Collapse
|
3
|
Fan X, Chen H, Xu C, Wang Y, Yin P, Li M, Tang Z, Jiang F, Wei W, Song J, Li G, Zhong D. Inhibiting Sphingosine 1-Phosphate Receptor Subtype 3 Attenuates Brain Damage During Ischemia-Reperfusion Injury by Regulating nNOS/NO and Oxidative Stress. Front Neurosci 2022; 16:838621. [PMID: 35242008 PMCID: PMC8886115 DOI: 10.3389/fnins.2022.838621] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/17/2022] [Indexed: 11/19/2022] Open
Abstract
Background Ischemic stroke (IS) is a common disease endangering human life and health. Cerebral ischemia triggers a series of complex harmful events, including excitotoxicity, inflammation and cell death, as well as increased nitric oxide production through the activation of nitric oxide synthase (NOS). Oxidative stress plays a major role in cerebral ischemia and reperfusion. Sphingosine 1-phosphate receptor subtype 3 (S1PR3), a member of S1P’s G protein-coupled receptors S1PR1-S1PR5, is involved in a variety of biological effects in the body, and its role in regulating oxidative stress during cerebral ischemia and reperfusion is still unclear. Methods Transient middle cerebral artery occlusion (tMCAO) mice were selected as the brain ischemia–reperfusion (I/R) injury model. Male C57/BL6 mice were treated with or without a selective S1PR3 inhibition after tMCAO, and changes in infarct volume, Nissl staining, hematoxylin-eosin (H&E) staining and NOS protein, nitric oxide (NO), superoxide dismutase (SOD), and malondialdehyde (MDA) content after tMCAO were observed. Results In the cerebral ischemia–reperfusion model, inhibition of S1PR3 improved the infarct volume and neuronal damage in mice after tMCAO. Similarly, inhibition of S1PR3 can reduce the expression of NO synthase subtype neuronal NOS (nNOS) and reduce the production of NO after cerebral ischemia. After cerebral ischemia and reperfusion, the oxidative stress response was enhanced, and after the administration of the S1PR3 inhibitor, the SOD content increased and the MDA content decreased, indicating that S1PR3 plays an important role in regulating oxidative stress response. Conclusion Inhibiting S1PR3 attenuates brain damage during I/R injury by regulating nNOS/NO and oxidative stress, which provides a potential new therapeutic target and mechanism for the clinical treatment of IS.
Collapse
Affiliation(s)
- Xuehui Fan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongping Chen
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chen Xu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yingju Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pengqi Yin
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meng Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhanbin Tang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fangchao Jiang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wan Wei
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jihe Song
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Di Zhong
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
4
|
Di Pietro P, Carrizzo A, Sommella E, Oliveti M, Iacoviello L, Di Castelnuovo A, Acernese F, Damato A, De Lucia M, Merciai F, Iesu P, Venturini E, Izzo R, Trimarco V, Ciccarelli M, Giugliano G, Carnevale R, Cammisotto V, Migliarino S, Virtuoso N, Strianese A, Izzo V, Campiglia P, Ciaglia E, Levkau B, Puca AA, Vecchione C. Targeting the ASMase/S1P pathway protects from sortilin-evoked vascular damage in hypertension. J Clin Invest 2022; 132:146343. [PMID: 35104805 PMCID: PMC8803332 DOI: 10.1172/jci146343] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 11/24/2021] [Indexed: 12/24/2022] Open
Abstract
Sortilin has been positively correlated with vascular disorders in humans. No study has yet evaluated the possible direct effect of sortilin on vascular function. We used pharmacological and genetic approaches coupled with study of murine and human samples to unravel the mechanisms recruited by sortilin in the vascular system. Sortilin induced endothelial dysfunction of mesenteric arteries through NADPH oxidase 2 (NOX2) isoform activation, dysfunction that was prevented by knockdown of acid sphingomyelinase (ASMase) or sphingosine kinase 1. In vivo, recombinant sortilin administration induced arterial hypertension in WT mice. In contrast, genetic deletion of sphingosine-1-phosphate receptor 3 (S1P3) and gp91phox/NOX2 resulted in preservation of endothelial function and blood pressure homeostasis after 14 days of systemic sortilin administration. Translating these research findings into the clinical setting, we detected elevated sortilin levels in hypertensive patients with endothelial dysfunction. Furthermore, in a population-based cohort of 270 subjects, we showed increased plasma ASMase activity and increased plasma levels of sortilin, S1P, and soluble NOX2-derived peptide (sNOX2-dp) in hypertensive subjects, and the increase was more pronounced in hypertensive subjects with uncontrolled blood pressure. Our studies reveal what we believe is a previously unrecognized role of sortilin in the impairment of vascular function and in blood pressure homeostasis and suggest the potential of sortilin and its mediators as biomarkers for the prediction of vascular dysfunction and high blood pressure.
Collapse
Affiliation(s)
- Paola Di Pietro
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy.,Department of Vascular Physiopathology, IRCCS Neuromed, Pozzilli, Italy
| | - Eduardo Sommella
- Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano, Italy
| | - Marco Oliveti
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Licia Iacoviello
- Department of Medicine and Surgery, Research Center in Epidemiology and Preventive Medicine (EPIMED), University of Insubria, Varese, Italy.,Department of Epidemiology and Prevention, IRCCS Neuromed, Pozzilli, Italy
| | | | - Fausto Acernese
- Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano, Italy
| | - Antonio Damato
- Department of Vascular Physiopathology, IRCCS Neuromed, Pozzilli, Italy
| | | | - Fabrizio Merciai
- Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano, Italy.,PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, Italy
| | - Paola Iesu
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | | | - Raffaele Izzo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Valentina Trimarco
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Giuseppe Giugliano
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Roberto Carnevale
- Mediterranea Cardiocentro, Naples, Italy.,Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Vittoria Cammisotto
- Department of General Surgery and Surgical Speciality Paride Stefanini, Sapienza University of Rome, Rome, Italy
| | - Serena Migliarino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Nicola Virtuoso
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Andrea Strianese
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Viviana Izzo
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Pietro Campiglia
- Department of Pharmacy, School of Pharmacy, University of Salerno, Fisciano, Italy.,European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | - Elena Ciaglia
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy
| | - Bodo Levkau
- Institute for Molecular Medicine III, Heinrich-Heine-University, Medical Faculty, Cardiovascular Research Institute Düsseldorf (CARID), Düsseldorf, Germany
| | - Annibale A Puca
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy.,Ageing Unit, IRCCS MultiMedica, Milan, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana" University of Salerno, Baronissi, Italy.,Department of Vascular Physiopathology, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
5
|
Zhang Y, Zhao Y, Ao X, Yu W, Zhang L, Wang Y, Chang W. The Role of Non-coding RNAs in Alzheimer's Disease: From Regulated Mechanism to Therapeutic Targets and Diagnostic Biomarkers. Front Aging Neurosci 2021; 13:654978. [PMID: 34276336 PMCID: PMC8283767 DOI: 10.3389/fnagi.2021.654978] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/11/2021] [Indexed: 01/05/2023] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder. AD is characterized by the production and aggregation of beta-amyloid (Aβ) peptides, hyperphosphorylated tau proteins that form neurofibrillary tangles (NFTs), and subsequent neuroinflammation, synaptic dysfunction, autophagy and oxidative stress. Non-coding RNAs (ncRNAs) can be used as potential therapeutic targets and biomarkers due to their vital regulatory roles in multiple biological processes involved in disease development. The involvement of ncRNAs in the pathogenesis of AD has been increasingly recognized. Here, we review the ncRNAs implicated in AD and elaborate on their main regulatory pathways, which might have contributions for discovering novel therapeutic targets and drugs for AD.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yanfang Zhao
- Institute of Biomedical Research, School for Life Science, Shandong University of Technology, Zibo, China
| | - Xiang Ao
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Wanpeng Yu
- School of Basic Medical Sciences, Qingdao University, Qingdao, China
| | - Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yu Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Lysophospholipids in Lung Inflammatory Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:373-391. [PMID: 33788203 DOI: 10.1007/978-3-030-63046-1_20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The lysophospholipids (LPLs) belong to a group of bioactive lipids that play pivotal roles in several physiological and pathological processes. LPLs are derivatives of phospholipids and consist of a single hydrophobic fatty acid chain, a hydrophilic head, and a phosphate group with or without a large molecule attached. Among the LPLs, lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are the simplest, and have been shown to be involved in lung inflammatory symptoms and diseases such as acute lung injury, asthma, and chronic obstructive pulmonary diseases. G protein-coupled receptors (GPCRs) mediate LPA and S1P signaling. In this chapter, we will discuss on the role of LPA, S1P, their metabolizing enzymes, inhibitors or agonists of their receptors, and their GPCR-mediated signaling in lung inflammatory symptoms and diseases, focusing specially on acute respiratory distress syndrome, asthma, and chronic obstructive pulmonary disease.
Collapse
|
7
|
Ha AW, Sudhadevi T, Ebenezer DL, Fu P, Berdyshev EV, Ackerman SJ, Natarajan V, Harijith A. Neonatal therapy with PF543, a sphingosine kinase 1 inhibitor, ameliorates hyperoxia-induced airway remodeling in a murine model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2020; 319:L497-L512. [PMID: 32697651 DOI: 10.1152/ajplung.00169.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hyperoxia (HO)-induced lung injury contributes to bronchopulmonary dysplasia (BPD) in preterm newborns. Intractable wheezing seen in BPD survivors is associated with airway remodeling (AWRM). Sphingosine kinase 1 (SPHK1)/sphingosine-1-phosphate (S1P) signaling promotes HO-mediated neonatal BPD; however, its role in the sequela of AWRM is not known. We noted an increased concentration of S1P in tracheal aspirates of neonatal infants with severe BPD, and earlier, demonstrated that Sphk1-/- mice showed protection against HO-induced BPD. The role of SPHK1/S1P in promoting AWRM following exposure of neonates to HO was investigated in a murine model. Therapy using PF543, the specific SPHK1 inhibitor, during neonatal HO reduced alveolar simplification followed by reduced AWRM in adult mice. This was associated with reduced airway hyperreactivity to intravenous methacholine. Neonatal HO exposure was associated with increased expression of SPHK1 in lung tissue of adult mice, which was reduced with PF543 therapy in the neonatal stage. This was accompanied by amelioration of HO-induced reduction of E-cadherin in airway epithelium. This may be suggestive of arrested partial epithelial mesenchymal transition (EMT) induced by HO. In vitro studies using human primary airway epithelial cells (HAEpCs) showed that SPHK1 inhibition or deletion restored HO-induced reduction in E-cadherin and reduced formation of mitochondrial reactive oxygen species (mtROS). Blocking mtROS with MitoTempo attenuated HO-induced partial EMT of HAEpCs. These results collectively support a therapeutic role for PF543 in preventing HO-induced BPD in neonates and the long-term sequela of AWRM, thus conferring a long-term protection resulting in improved lung development and function.
Collapse
Affiliation(s)
- Alison W Ha
- Department of Biochemistry, University of Illinois, Chicago, Illinois
| | - Tara Sudhadevi
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - David L Ebenezer
- Department of Pharmacology, University of Illinois, Chicago, Illinois
| | - Panfeng Fu
- Department of Pharmacology, University of Illinois, Chicago, Illinois
| | | | - Steven J Ackerman
- Department of Biochemistry, University of Illinois, Chicago, Illinois
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois, Chicago, Illinois.,Department of Medicine, University of Illinois, Chicago, Illinois
| | - Anantha Harijith
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
8
|
Chen Y, Wang JC, Yang CM, Fan Q, Zheng J, Liu H. Positive acceleration adaptive training attenuates gastric ischemia-reperfusion injury through COX-2 and PGE2 expression. Exp Ther Med 2019; 17:2901-2906. [PMID: 30930978 PMCID: PMC6425289 DOI: 10.3892/etm.2019.7288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 03/02/2018] [Indexed: 12/19/2022] Open
Abstract
The mechanism involved in the effects of positive acceleration adaptive training (PAAT) on gastric ischemia-reperfusion injury (GI-RI) has not been fully characterized. The aim of the present study was to investigate the effects of PAAT in attenuating GI-RI in a rat model. The inflammatory factor and caspase-3 levels were measured using ELISA kits. A western blot assay was used to analyze tumor necrosis factor-α (TNF)-α, tumor necrosis factor receptor 1 (TNFR1), tumor necrosis factor-related apoptosis inducing ligand (TRAIL), death receptor (DR) 4, DR5, cyclooxygenase (COX)-2, COX-1 and prostaglandin E2 (PGE2) protein expression levels. It was revealed that PAAT could alleviate GI-RI and inflammatory factor levels in a rat model. PAAT suppressed TNF-α and TNFR1 protein expression levels, inhibited TRAIL, DR4, DR5, COX-2 and PGE2 protein expression levels; however, it did not have an effect on COX-1 protein expression in the model of GI-RI. The data indicated that the effects of PAAT attenuated GI-RI through the downregulation of COX-2 and PGE2 expression.
Collapse
Affiliation(s)
- Ying Chen
- Department of Gastroenterology and Research Center of Aeropathy, General Hospital of The Air Force, People's Liberation Army, Beijing 100036, P.R. China
| | - Jian-Chang Wang
- Department of Gastroenterology and Research Center of Aeropathy, General Hospital of The Air Force, People's Liberation Army, Beijing 100036, P.R. China
| | - Chun-Min Yang
- Department of Gastroenterology and Research Center of Aeropathy, General Hospital of The Air Force, People's Liberation Army, Beijing 100036, P.R. China
| | - Qin Fan
- Department of Gastroenterology and Research Center of Aeropathy, General Hospital of The Air Force, People's Liberation Army, Beijing 100036, P.R. China
| | - Jun Zheng
- Department of Gastroenterology and Research Center of Aeropathy, General Hospital of The Air Force, People's Liberation Army, Beijing 100036, P.R. China
| | - Hao Liu
- Department of Gastroenterology and Research Center of Aeropathy, General Hospital of The Air Force, People's Liberation Army, Beijing 100036, P.R. China
| |
Collapse
|
9
|
Wang Y, Feng F. Evaluation of the Hepatotoxicity of the Zhi-Zi-Hou-Po Decoction by Combining UPLC-Q-Exactive-MS-Based Metabolomics and HPLC-MS/MS-Based Geniposide Tissue Distribution. Molecules 2019; 24:E511. [PMID: 30708983 PMCID: PMC6384998 DOI: 10.3390/molecules24030511] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/24/2019] [Accepted: 01/27/2019] [Indexed: 12/20/2022] Open
Abstract
With traditional Chinese medicine (TCM) becoming widespread globally, its safety has increasingly become a concern, especially its hepatoxicity. For example, Gardenia jasminoides Ellis is a key ingredient in the Zhi-Zi-Hou-Po decoction (ZZHPD), which is a commonly-used clinically combined prescription of TCM that may induce hepatoxicity. However, the underlying toxicity mechanism of ZZHPD is not fully understood. In this study, a plasma metabolomics strategy was used to investigate the mechanism of ZZHPD-induced hepatotoxicity through profiling entire endogenous metabolites. Twenty-four Sprague-Dawley rats were randomly assigned into four groups, which were orally administered with 0.9% saline, as well as 2.7 g/kg/day, 8.1 g/kg/day, or 27 g/kg/day of ZZHPD for 30 consecutive days, respectively. Biochemical assay and metabolomics assay were used to detect serum and plasma samples, whilst histopathological assay was used for detecting liver tissues, and the geniposide distribution in tissues was simultaneously measured. The results showed that the concentration of 20 metabolites linked to amino acid, lipid, and bile acid metabolism had significant changes in the ZZHPD-treated rats. Moreover, toxic effects were aggravated with serum biochemical and histopathological examines in liver tissues as the dosage increased, which may be associated with the accumulation of geniposide in the liver as the dosage increased. Notably, our findings also demonstrated that the combined metabolomics strategy with tissue distribution had significant potential for elucidating the mechanistic complexity of the toxicity of TCM.
Collapse
Affiliation(s)
- Yunji Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| | - Fang Feng
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
10
|
Metformin Inhibits Migration and Invasion by Suppressing ROS Production and COX2 Expression in MDA-MB-231 Breast Cancer Cells. Int J Mol Sci 2018; 19:ijms19113692. [PMID: 30469399 PMCID: PMC6274682 DOI: 10.3390/ijms19113692] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/16/2018] [Accepted: 11/17/2018] [Indexed: 02/07/2023] Open
Abstract
Background: Several mechanisms of action have been proposed to explain the apparent antineoplastic functions of metformin, many of which are observed at high concentrations that may not be reflective of achievable tissue concentrations. We propose that metformin at low concentrations functions to inhibit ROS production and inflammatory signaling in breast cancer, thereby reducing metastasis. Methods: Using the highly invasive MDA-MB-231 breast carcinoma model, we ascertained the impact of metformin on cell viability by DNA content analysis and fluorescent dye exclusion. Migration and invasion assays were performed using a modified Boyden chamber assay and metastasis was ascertained using the chorioallantoic membrane (CAM) assay. PGE2 production was measured by Enzyme-Linked Immunosorbent Assay (ELISA). COX2 and ICAM1 levels were determined by flow cytometry immunoassay. Results: Metformin acutely decreased cell viability and caused G2 cell cycle arrest only at high concentrations (10 mM). At 100 µM, however, metformin reduced ICAM1 and COX2 expression, as well as reduced PGE2 production and endogenous mitochondrial ROS production while failing to significantly impact cell viability. Consequently, metformin inhibited migration, invasion in vitro and PGE2-dependent metastasis in CAM assays. Conclusion: At pharmacologically achievable concentrations, metformin does not drastically impact cell viability, but inhibits inflammatory signaling and metastatic progression in breast cancer cells.
Collapse
|
11
|
Liu CW, Lee TL, Chen YC, Liang CJ, Wang SH, Lue JH, Tsai JS, Lee SW, Chen SH, Yang YF, Chuang TY, Chen YL. PM 2.5-induced oxidative stress increases intercellular adhesion molecule-1 expression in lung epithelial cells through the IL-6/AKT/STAT3/NF-κB-dependent pathway. Part Fibre Toxicol 2018; 15:4. [PMID: 29329563 PMCID: PMC5767014 DOI: 10.1186/s12989-018-0240-x] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Epidemiological studies have shown that ambient air pollution is closely associated with increased respiratory inflammation and decreased lung function. Particulate matters (PMs) are major components of air pollution that damages lung cells. However, the mechanisms remain to be elucidated. This study examines the effects of PMs on intercellular adhesion molecule-1 (ICAM-1) expression and the related mechanisms in vitro and in vivo. RESULT The cytotoxicity, reactive oxygen species (ROS) generation, and monocyte adherence to A549 cells were more severely affected by treatment with O-PMs (organic solvent-extractable fraction of SRM1649b) than with W-PMs (water-soluble fraction of SRM1649b). We observed a significant increase in ICAM-1 expression by O-PMs, but not W-PMs. O-PMs also induced the phosphorylation of AKT, p65, and STAT3. Pretreating A549 cells with N-acetyl cysteine (NAC), an antioxidant, attenuated O-PMs-induced ROS generation, the phosphorylation of the mentioned kinases, and the expression of ICAM-1. Furthermore, an AKT inhibitor (LY294002), NF-κB inhibitor (BAY11-7082), and STAT3 inhibitor (Stattic) significantly down-regulated O-PMs-induced ICAM-1 expression as well as the adhesion of U937 cells to epithelial cells. Interleukin-6 (IL-6) was the most significantly changed cytokine in O-PMs-treated A549 cells according to the analysis of the cytokine antibody array. The IL-6 receptor inhibitor tocilizumab (TCZ) and small interfering RNA for IL-6 significantly reduced ICAM-1 secretion and expression as well as the reduction of the AKT, p65, and STAT3 phosphorylation in O-PMs-treated A549 cells. In addition, the intratracheal instillation of PMs significantly increased the levels of the ICAM-1 and IL-6 in lung tissues and plasma in WT mice, but not in IL-6 knockout mice. Pre-administration of NAC attenuated those PMs-induced adverse effects in WT mice. Furthermore, patients with chronic obstructive pulmonary disease (COPD) had higher plasma levels of ICAM-1 and IL-6 compared to healthy subjects. CONCLUSION These results suggest that PMs increase ICAM-1 expression in pulmonary epithelial cells in vitro and in vivo through the IL-6/AKT/STAT3/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chen-Wei Liu
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Sec 1, Ren-Ai Road, Taipei, Taiwan
| | - Tzu-Lin Lee
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Sec 1, Ren-Ai Road, Taipei, Taiwan
| | - Yu-Chen Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Sec 1, Ren-Ai Road, Taipei, Taiwan
| | - Chan-Jung Liang
- Lipid Science and Aging Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Lipid Biosciences, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shu-Huei Wang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Sec 1, Ren-Ai Road, Taipei, Taiwan
| | - June-Horng Lue
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Sec 1, Ren-Ai Road, Taipei, Taiwan
| | - Jaw-Shiun Tsai
- Department of Family Medicine, College of Medicine and Hospital, Taipei, Taiwan.,Center for Complementary and Integrated Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Wei Lee
- Department of Internal Medicine, Taoyuan General Hospital, Department of Health and Welfare, No.1492, Zhongshan Road, Taoyuan, Taiwan
| | - Shun-Hua Chen
- Department of Microbiology and Immunology, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Fan Yang
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Yi Chuang
- Department of Internal Medicine, Taoyuan General Hospital, Department of Health and Welfare, No.1492, Zhongshan Road, Taoyuan, Taiwan. .,Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Sec 1, Ren-Ai Road, Taipei, Taiwan.
| |
Collapse
|
12
|
Dietary and Endogenous Sphingolipid Metabolism in Chronic Inflammation. Nutrients 2017; 9:nu9111180. [PMID: 29143791 PMCID: PMC5707652 DOI: 10.3390/nu9111180] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/21/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022] Open
Abstract
Chronic inflammation is a common underlying factor in many major metabolic diseases afflicting Western societies. Sphingolipid metabolism is pivotal in the regulation of inflammatory signaling pathways. The regulation of sphingolipid metabolism is in turn influenced by inflammatory pathways. In this review, we provide an overview of sphingolipid metabolism in mammalian cells, including a description of sphingolipid structure, biosynthesis, turnover, and role in inflammatory signaling. Sphingolipid metabolites play distinct and complex roles in inflammatory signaling and will be discussed. We also review studies examining dietary sphingolipids and inflammation, derived from in vitro and rodent models, as well as human clinical trials. Dietary sphingolipids appear to influence inflammation-related chronic diseases through inhibiting intestinal lipid absorption, altering gut microbiota, activation of anti-inflammatory nuclear receptors, and neutralizing responses to inflammatory stimuli. The anti-inflammatory effects observed with consuming dietary sphingolipids are in contrast to the observation that most cellular sphingolipids play roles in augmenting inflammatory signaling. The relationship between dietary sphingolipids and low-grade chronic inflammation in metabolic disorders is complex and appears to depend on sphingolipid structure, digestion, and metabolic state of the organism. Further research is necessary to confirm the reported anti-inflammatory effects of dietary sphingolipids and delineate their impacts on endogenous sphingolipid metabolism.
Collapse
|
13
|
Sphingosine-1-phosphate is involved in inflammatory reactions in patients with Graves’ orbitopathy. Inflamm Res 2017; 66:535-545. [DOI: 10.1007/s00011-017-1037-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/08/2017] [Accepted: 03/13/2017] [Indexed: 01/06/2023] Open
|