1
|
Wang B, Wu X, Cheng J, Ye J, Zhu H, Liu X. Regulatory role of S1P and its receptors in sepsis-induced liver injury. Front Immunol 2025; 16:1489015. [PMID: 39935473 PMCID: PMC11811114 DOI: 10.3389/fimmu.2025.1489015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
As an immune and metabolic organ, the liver affects the progression and prognosis of sepsis. Despite the severe adverse effects of sepsis liver injury on the body, treatment options remain limited. Sphingosine-1-phosphate (S1P) is a widely distributed lipid signaling molecule that binds to five sphingosine-1-phosphate receptors (S1PR) to regulate downstream signaling pathways involved in the pathophysiological processes of sepsis, including endothelial permeability, cytokine release, and vascular tone. This review summarizes current research on the role of S1P in normal liver biology and describes the mechanisms by which changes in S1P/S1PR affect the development of liver-related diseases. At the same time, the pathological processes underlying liver injury, as evidenced by clinical manifestations during sepsis, were comprehensively reviewed. This paper focused on the mechanistic pathways through which S1P and its receptors modulate immunity, bile acid metabolism, and liver-intestinal circulation in septic liver injury. Finally, the relationships between S1P and its receptors with liver inflammation and metabolism and the use of related drugs for the treatment of liver injury were examined. By elucidating the role of S1P and its receptor in the pathogenesis of sepsis liver injury, this review established a molecular targeting framework, providing novel insights into clinical and drug development.
Collapse
Affiliation(s)
- Bin Wang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiaoyu Wu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jiangfeng Cheng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junming Ye
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Clinical College, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Hongquan Zhu
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiaofeng Liu
- Clinical College, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Emergency, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
2
|
Smirne C, Crobu MG, Landi I, Vercellino N, Apostolo D, Pinato DJ, Vincenzi F, Minisini R, Tonello S, D’Onghia D, Ottobrelli A, Martini S, Bracco C, Fenoglio LM, Campanini M, Berton AM, Ciancio A, Pirisi M. Chronic Hepatitis C Infection Treated with Direct-Acting Antiviral Agents and Occurrence/Recurrence of Hepatocellular Carcinoma: Does It Still Matter? Viruses 2024; 16:1899. [PMID: 39772206 PMCID: PMC11680226 DOI: 10.3390/v16121899] [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: 10/09/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/03/2025] Open
Abstract
Hepatitis C virus (HCV) infection is a significant risk factor for liver cirrhosis and hepatocellular carcinoma (HCC). Traditionally, the primary prevention strategy for HCV-associated HCC has focused on removing infection through antiviral regimes. Currently, highly effective direct-acting antivirals (DAAs) offer extraordinary success across all patient categories, including cirrhotics. Despite these advancements, recent studies have reported that even after sustained virologic response (SVR), individuals with advanced liver disease/cirrhosis at the time of DAA treatment may still face risks of HCC occurrence or recurrence. Based on this premise, this review tries to shed light on the multiple mechanisms that establish a tumorigenic environment, first, during chronic HCV infection and then, after eventual viral eradication by DAAs. Furthermore, it reviews evidence reported by recent observational studies stating that the use of DAAs is not associated with an increased risk of HCC development but rather, with a significantly lower chance of liver cancer compared with DAA-untreated patients. In addition, it seeks to provide some practical guidance for clinicians, helping them to manage HCC surveillance of patients who have achieved SVR with DAAs.
Collapse
Affiliation(s)
- Carlo Smirne
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (N.V.); (D.A.); (D.J.P.); (F.V.); (R.M.); (S.T.); (D.D.); (M.C.); (M.P.)
- Internal Medicine Unit, Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Maria Grazia Crobu
- Laboratory of Molecular Virology, Maggiore della Carità Hospital, 28100 Novara, Italy;
- Clinical Biochemistry Laboratory, City of Health and Science University Hospital, 10126 Turin, Italy
| | - Irene Landi
- Emergency Medicine Department, Michele e Pietro Ferrero Hospital, 12060 Verduno, Italy;
| | - Nicole Vercellino
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (N.V.); (D.A.); (D.J.P.); (F.V.); (R.M.); (S.T.); (D.D.); (M.C.); (M.P.)
| | - Daria Apostolo
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (N.V.); (D.A.); (D.J.P.); (F.V.); (R.M.); (S.T.); (D.D.); (M.C.); (M.P.)
| | - David James Pinato
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (N.V.); (D.A.); (D.J.P.); (F.V.); (R.M.); (S.T.); (D.D.); (M.C.); (M.P.)
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, London SW7 2AZ, UK
| | - Federica Vincenzi
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (N.V.); (D.A.); (D.J.P.); (F.V.); (R.M.); (S.T.); (D.D.); (M.C.); (M.P.)
| | - Rosalba Minisini
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (N.V.); (D.A.); (D.J.P.); (F.V.); (R.M.); (S.T.); (D.D.); (M.C.); (M.P.)
| | - Stelvio Tonello
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (N.V.); (D.A.); (D.J.P.); (F.V.); (R.M.); (S.T.); (D.D.); (M.C.); (M.P.)
| | - Davide D’Onghia
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (N.V.); (D.A.); (D.J.P.); (F.V.); (R.M.); (S.T.); (D.D.); (M.C.); (M.P.)
| | - Antonio Ottobrelli
- Gastroenterology Unit, City of Health and Science University Hospital, 10126 Turin, Italy; (A.O.); (S.M.); (A.C.)
| | - Silvia Martini
- Gastroenterology Unit, City of Health and Science University Hospital, 10126 Turin, Italy; (A.O.); (S.M.); (A.C.)
| | - Christian Bracco
- Department of Internal Medicine, Santa Croce e Carle Hospital, 12100 Cuneo, Italy; (C.B.); (L.M.F.)
| | - Luigi Maria Fenoglio
- Department of Internal Medicine, Santa Croce e Carle Hospital, 12100 Cuneo, Italy; (C.B.); (L.M.F.)
| | - Mauro Campanini
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (N.V.); (D.A.); (D.J.P.); (F.V.); (R.M.); (S.T.); (D.D.); (M.C.); (M.P.)
- Internal Medicine Unit, Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Alessandro Maria Berton
- Division of Endocrinology, Diabetes and Metabolism, City of Health and Science University Hospital, 10126 Turin, Italy;
| | - Alessia Ciancio
- Gastroenterology Unit, City of Health and Science University Hospital, 10126 Turin, Italy; (A.O.); (S.M.); (A.C.)
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Mario Pirisi
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (N.V.); (D.A.); (D.J.P.); (F.V.); (R.M.); (S.T.); (D.D.); (M.C.); (M.P.)
- Internal Medicine Unit, Maggiore della Carità Hospital, 28100 Novara, Italy
| |
Collapse
|
3
|
Gautam J, Aggarwal H, Kumari D, Gupta SK, Kumar Y, Dikshit M. A methionine-choline-deficient diet induces nonalcoholic steatohepatitis and alters the lipidome, metabolome, and gut microbiome profile in the C57BL/6J mouse. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159545. [PMID: 39089643 DOI: 10.1016/j.bbalip.2024.159545] [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: 02/12/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
The methionine-choline-deficient (MCD) diet-induced non-alcoholic steatohepatitis (NASH) in mice is a well-established model. Our study aims to elucidate the factors influencing liver pathology in the MCD mouse model by examining physiological, biochemical, and molecular changes using histology, molecular techniques, and OMICS approaches (lipidomics, metabolomics, and metagenomics). Male C57BL/6J mice were fed a standard chow diet, a methionine-choline-sufficient (MCS) diet, or an MCD diet for 10 weeks. The MCD diet resulted in reduced body weight and fat mass, along with decreased plasma triglyceride, cholesterol, glucose, and insulin levels. However, it notably induced steatosis, inflammation, and alterations in gene expression associated with lipogenesis, inflammation, fibrosis, and the synthesis of apolipoproteins, sphingolipids, ceramides, and carboxylesterases. Lipid analysis revealed significant changes in plasma and tissues: most ceramide non-hydroxy-sphingosine lipids significantly decreased in the liver and plasma but increased in the adipose tissue of MCD diet-fed animals. Oxidized glycerophospholipids mostly increased in the liver but decreased in the adipose tissue of the MCD diet-fed group. The gut microbiome of the MCD diet-fed group showed an increase in Firmicutes and a decrease in Bacteroidetes and Actinobacteria. Metabolomic profiling demonstrated that the MCD diet significantly altered amino acid biosynthesis, metabolism, and nucleic acid metabolism pathways in plasma, liver, fecal, and cecal samples. LC-MS data indicated higher total plasma bile acid intensity and reduced fecal glycohyodeoxycholic acid intensity in the MCD diet group. This study demonstrates that although the MCD diet induces hepatic steatosis, the mechanisms underlying NASH in this model differ from those in human NASH pathology.
Collapse
Affiliation(s)
- Jyoti Gautam
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Hobby Aggarwal
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Deepika Kumari
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Sonu Kumar Gupta
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India
| | - Yashwant Kumar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| | - Madhu Dikshit
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad 121001, Haryana, India.
| |
Collapse
|
4
|
Zhang L, Song YH, Liu J, Zhao YX, Zhou RR, Xu JC, He J, Lu YL, Gan WJ, Lu XS, Li M, Zhou P, Wang L, Han QZ. Hepatitis B Virus Increases SphK1-S1P Synthesis by Promoting the Availability of the Transcription Factor USF1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1499-1507. [PMID: 39400236 PMCID: PMC11533153 DOI: 10.4049/jimmunol.2400088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024]
Abstract
Hepatitis B virus (HBV) is the most common chronic viral infection globally, affecting ∼360 million people and causing about 1 million deaths annually due to end-stage liver disease or hepatocellular carcinoma. Current antiviral treatments rarely achieve a functional cure for chronic hepatitis B, highlighting the need for improved monitoring and intervention strategies. This study explores the role of the sphingosine kinase 1 (SphK1)-sphingosine-1-phosphate (S1P) axis in HBV-related liver injury. We investigated the association between serum S1P concentration and HBV DNA levels in chronic hepatitis B patients, finding a significant positive correlation. Additionally, SphK1 was elevated in liver tissues of HBV-positive hepatocellular carcinoma patients, particularly in HBsAg-positive regions. HBV infection models in HepG2-sodium taurocholate cotransporting polypeptide cells confirmed that HBV enhances SphK1 expression and S1P production. Inhibition of HBV replication through antiviral agents and the CRISPR-Cas9 system reduced SphK1 and S1P levels. Further, we identified the transcription factor USF1 as a key regulator of SphK1 expression during HBV infection. USF1 binds to the SphK1 promoter, increasing its transcriptional activity, and is upregulated in response to HBV infection. In vivo studies in mice demonstrated that HBV exposure promotes the expression of USF1 and SphK1-S1P. These findings suggest that the SphK1-S1P axis, regulated by HBV-induced USF1, could serve as a potential biomarker and therapeutic target for HBV-related liver injury.
Collapse
Affiliation(s)
- Lu Zhang
- Center of Clinical Laboratory and Translational Medicine, Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People’s Republic of China
- Department of Laboratory Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ya-Hui Song
- Center of Clinical Laboratory and Translational Medicine, Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People’s Republic of China
| | - Juan Liu
- Center of Clinical Laboratory and Translational Medicine, Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People’s Republic of China
| | - Yin-Xia Zhao
- Central Laboratory, Shanghai Xuhui Central Hospital/Zhongshan–Xuhui Hospital, Fudan University, Shanghai, China
| | - Ruo-Ran Zhou
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu, People’s Republic of China
| | - Jun-Chi Xu
- Fifth People’s Hospital of Suzhou, Suzhou, People’s Republic of China
| | - Jun He
- Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, People’s Republic of China
| | - You-Li Lu
- Central Laboratory, Shanghai Xuhui Central Hospital/Zhongshan–Xuhui Hospital, Fudan University, Shanghai, China
| | - Wen-Juan Gan
- Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People’s Republic of China
| | - Xing-Sheng Lu
- Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People’s Republic of China
| | - Min Li
- Institute of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, People’s Republic of China
| | - Peng Zhou
- Center of Clinical Laboratory and Translational Medicine, Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People’s Republic of China
| | - Lin Wang
- Center of Clinical Laboratory and Translational Medicine, Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People’s Republic of China
| | - Qing-Zhen Han
- Center of Clinical Laboratory and Translational Medicine, Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, Jiangsu, People’s Republic of China
| |
Collapse
|
5
|
Lkham-Erdene B, Choijookhuu N, Kubota T, Uto T, Mitoma S, Shirouzu S, Ishizuka T, Kai K, Higuchi K, Mo Aung K, Batmunkh JE, Sato K, Hishikawa Y. Effect of Hepatic Lipid Overload on Accelerated Hepatocyte Proliferation Promoted by HGF Expression via the SphK1/S1PR2 Pathway in MCD-diet Mouse Partial Hepatectomy. Acta Histochem Cytochem 2024; 57:175-188. [PMID: 39552932 PMCID: PMC11565223 DOI: 10.1267/ahc.24-00046] [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: 09/01/2024] [Accepted: 09/17/2024] [Indexed: 11/19/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is becoming a major health problem worldwide. Liver regeneration is crucial for restoring liver function, and is regulated by extraordinary complex process, involving numerous factors under both physiologic and pathologic conditions. Sphingosine-1-phosphate (S1P), a bioactive sphingolipid synthesized by sphingosine kinase 1 (SphK1), plays an important role in liver function through S1P receptors (S1PRs)-expressing cells. In this study, we investigated the effect of lipid overload on hepatocyte proliferation in a mouse hepatic steatosis model induced by feeding a methionine- and choline-deficient (MCD) diet. After 50% partial hepatectomy (PHx), liver tissues were sampled at various timepoints and then analyzed by immunohistochemistry, oil Red-O staining, quantitative-polymerase chain reaction (qPCR), and flow cytometry. In mice fed the MCD-diet, significantly exacerbated hepatic steatosis and accelerated liver regeneration were observed. After PHx, hepatocyte proliferation peaked at 48 and 36 hr in the liver of chow- and MCD-diet fed mice, respectively. By contrast, increased expression of S1PR2 was observed in hepatic neutrophils and macrophages of MCD-diet fed mice. Flow cytometry and qPCR experiments demonstrated that levels of HGF and FGF2 released by neutrophils and macrophages were significantly higher in MCD-diet fed mice. In conclusion, hepatic lipid overload recruits Kupffer cells and neutrophils that release HGF and FGF2 via SphK1/S1PR2 activation to accelerate hepatocyte proliferation.
Collapse
Affiliation(s)
- Baljinnyam Lkham-Erdene
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
- Thoracic surgery department, National Cancer Center, Ulaanbaatar, Mongolia
| | - Narantsog Choijookhuu
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
- Department of Pathology and Forensic Medicine, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Toshiki Kubota
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| | - Tomofumi Uto
- Division of Immunology, Department of Infectious diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| | - Shuya Mitoma
- Division of Immunology, Department of Infectious diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| | - Shinichiro Shirouzu
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| | - Takumi Ishizuka
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| | - Kengo Kai
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| | - Kazuhiro Higuchi
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
- Department of Surgery, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| | - Kham Mo Aung
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| | - Jargal-Erdene Batmunkh
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| | - Katsuaki Sato
- Division of Immunology, Department of Infectious diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| | - Yoshitaka Hishikawa
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889–1692, Japan
| |
Collapse
|
6
|
Pérez-Cova M, Bedia C, Checa A, Meister I, Tauler R, Wheelock CE, Jaumot J. Metabolomic and sphingolipidomic profiling of human hepatoma cells exposed to widely used pharmaceuticals. J Pharm Biomed Anal 2024; 249:116378. [PMID: 39074424 DOI: 10.1016/j.jpba.2024.116378] [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: 04/11/2024] [Revised: 06/08/2024] [Accepted: 07/21/2024] [Indexed: 07/31/2024]
Abstract
Pharmaceutical compounds have become one of the main contaminants of emerging concern (CECs) due to their high usage and increased release into the environment. This study aims to assess the effects caused by three widely consumed hepatotoxic pharmaceutical compounds: an antibiotic (amoxicillin), an antiepileptic (carbamazepine), and an antidepressant (trazodone), on human health when indirectly exposed to toxicologically relevant concentrations (30, 15, and 7.5 μM for amoxicillin and carbamazepine, and 4, 2, and 1 μM for trazodone). A combination of semi-targeted metabolomic and targeted sphingolipid analyses was chosen to unravel the metabolic alterations in human hepatic cells exposed to these CECs at three concentrations for 24 h. HepG2 hepatoma cells were encapsulated in sodium alginate spheroids to improve the physiological relevance of this in vitro approach. Statistical analysis was used to identify the most affected metabolites and sphingolipids for each drug exposure. The results revealed small but significant changes in response to carbamazepine and trazodone exposures, affecting sphingolipid, glycerophospholipid precursors, and amino acid metabolism. Under both drug treatments, a decrease in various ceramide species (related to cell signaling) was observed, along with reduced taurine levels (related to the biosynthesis of bile acid conjugates) and carnitine levels (suggesting an impact on energy production). These and other drug-specific changes indicate that cellular functions in liver cells might be altered under low doses of these CECs, potentially affecting the health of other organs.
Collapse
Affiliation(s)
- Miriam Pérez-Cova
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain; Department of Chemical Engineering and Analytical Chemistry, University of Barcelona, Diagonal 647, Barcelona, Barcelona E08028, Spain
| | - Carmen Bedia
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain
| | - Antonio Checa
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Isabel Meister
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden; Gunma University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Romà Tauler
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain
| | - Craig E Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden; Gunma University Initiative for Advanced Research (GIAR), Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan; Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm 141-86, Sweden
| | - Joaquim Jaumot
- Department of Environmental Chemistry, IDAEA-CSIC, Jordi Girona 18-26, Barcelona E08034, Spain.
| |
Collapse
|
7
|
Yang C, Lv F, Yang J, Ding D, Cui L, Han Y. Surveillance and management of hepatocellular carcinoma after treatment of hepatitis C with direct-acting antiviral drugs. Ann Hepatol 2024; 30:101582. [PMID: 39276980 DOI: 10.1016/j.aohep.2024.101582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/11/2024] [Accepted: 08/15/2024] [Indexed: 09/17/2024]
Abstract
Hepatitis C virus (HCV) belongs to the Flaviviridae family, and is a single-stranded RNA virus with positive polarity. It is the primary cause of hepatocellular carcinoma (HCC) worldwide. The treatment of HCV has entered a new era with the advent of direct-acting antiviral drugs (DAAs) and is associated with cure rates of more than 95 %, making HCV the only curable viral disease. The successful treatment of chronic hepatitis C has greatly reduced, but not eliminated, the risk of HCC. Certain individuals, especially those with cirrhosis already present, remain vulnerable to HCC after achieving a sustained virological response (SVR). This article systematically reviews the recent studies on the risk and mechanisms of HCC development after HCV viral cure, the screening and predictive value of biological markers, and patient surveillance. Factors such as older age, diabetes, hepatic fat accumulation, alcohol use, and lack of fibrosis reversal are linked to increased HCC risk after HCV cure. The mechanism of HCC development after DAAs treatment remains unclear, but the possible mechanisms include immune cell dysfunction during HCV infection, cytokine network imbalance, epigenetic alterations, and host factors. Several biological markers and risk prediction models have been used to monitor the risk of HCC in CHC patients who have achieved SVR, but most still require validation and standardization. The implementation of risk-stratified surveillance programs is becoming urgent from a cost-effective point of view, but the availability of validated biomarkers to predict HCC in cured patients remains an unmet clinical need. Additionally, managing CHC patients who achieve SVR is becoming a growing challenge as an increasing number of HCV patients are cured.
Collapse
Affiliation(s)
- Caiyun Yang
- Xi'an Medical University, Xi'an 710021, Shannxi, PR China; National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, PR China
| | - Fengxiang Lv
- Xi'an Medical University, Xi'an 710021, Shannxi, PR China; National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, PR China
| | - Jiaqi Yang
- National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, PR China
| | - Dawei Ding
- National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, PR China
| | - Lina Cui
- National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, PR China.
| | - Ying Han
- National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Xijing Hospital, Air Force Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
8
|
Jia W, Yuan J, Zhang J, Li S, Lin W, Cheng B. Bioactive sphingolipids as emerging targets for signal transduction in cancer development. Biochim Biophys Acta Rev Cancer 2024; 1879:189176. [PMID: 39233263 DOI: 10.1016/j.bbcan.2024.189176] [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: 06/06/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Sphingolipids, crucial components of cellular membranes, play a vital role in maintaining cellular structure and signaling integrity. Disruptions in sphingolipid metabolism are increasingly implicated in cancer development. Key bioactive sphingolipids, such as ceramides, sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), and glycosphingolipids, profoundly impact tumor biology. They influence the behavior of tumor cells, stromal cells, and immune cells, affecting tumor aggressiveness, angiogenesis, immune modulation, and extracellular matrix remodeling. Furthermore, abnormal expression of sphingolipids and their metabolizing enzymes modulates the secretion of tumor-derived extracellular vesicles (TDEs), which are key players in creating an immunosuppressive tumor microenvironment, remodeling the extracellular matrix, and facilitating oncogenic signaling within in situ tumors and distant pre-metastatic niches (PMNs). Understanding the role of sphingolipids in the biogenesis of tumor-derived extracellular vesicles (TDEs) and their bioactive contents can pave the way for new biomarkers in cancer diagnosis and prognosis, ultimately enhancing comprehensive tumor treatment strategies.
Collapse
Affiliation(s)
- Wentao Jia
- Department of General Practice, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China
| | - Jiaying Yuan
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jinbo Zhang
- Department of Pharmacy, Tianjin Rehabilitation and Recuperation Center, Joint Logistics Support Force, Tianjin 300000, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Wanfu Lin
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| |
Collapse
|
9
|
Boccatonda A, Del Cane L, Marola L, D’Ardes D, Lessiani G, di Gregorio N, Ferri C, Cipollone F, Serra C, Santilli F, Piscaglia F. Platelet, Antiplatelet Therapy and Metabolic Dysfunction-Associated Steatotic Liver Disease: A Narrative Review. Life (Basel) 2024; 14:473. [PMID: 38672744 PMCID: PMC11051088 DOI: 10.3390/life14040473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is not only related to traditional cardiovascular risk factors like type 2 diabetes mellitus and obesity, but it is also an independent risk factor for the development of cardiovascular disease. MASLD has been shown to be independently related to endothelial dysfunction and atherosclerosis. MASLD is characterized by a chronic proinflammatory response that, in turn, may induce a prothrombotic state. Several mechanisms such as endothelial and platelet dysfunction, changes in the coagulative factors, lower fibrinolytic activity can contribute to induce the prothrombotic state. Platelets are players and addresses of metabolic dysregulation; obesity and insulin resistance are related to platelet hyperactivation. Furthermore, platelets can exert a direct effect on liver cells, particularly through the release of mediators from granules. Growing data in literature support the use of antiplatelet agent as a treatment for MASLD. The use of antiplatelets drugs seems to exert beneficial effects on hepatocellular carcinoma prevention in patients with MASLD, since platelets contribute to fibrosis progression and cancer development. This review aims to summarize the main data on the role of platelets in the pathogenesis of MASLD and its main complications such as cardiovascular events and the development of liver fibrosis. Furthermore, we will examine the role of antiplatelet therapy not only in the prevention and treatment of cardiovascular events but also as a possible anti-fibrotic and anti-tumor agent.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Internal Medicine, Bentivoglio Hospital, AUSL Bologna, 40010 Bentivoglio, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
| | - Lorenza Del Cane
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Lara Marola
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Damiano D’Ardes
- Institute of “Clinica Medica”, Department of Medicine and Aging Science, “G. D’Annunzio” University of Chieti, 66100 Chieti, Italy (F.C.)
| | | | - Nicoletta di Gregorio
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Claudio Ferri
- Nephrology Unit, Department of Life, Health & Environmental Sciences and Internal Medicine, University of L’Aquila, ASL Avezzano-Sulmona-L’Aquila, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.D.C.); (L.M.); (N.d.G.); (C.F.)
| | - Francesco Cipollone
- Institute of “Clinica Medica”, Department of Medicine and Aging Science, “G. D’Annunzio” University of Chieti, 66100 Chieti, Italy (F.C.)
| | - Carla Serra
- Interventional, Diagnostic and Therapeutic Ultrasound Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Francesca Santilli
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology, University of Chieti, 66100 Chieti, Italy;
| | - Fabio Piscaglia
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
10
|
Alkafaas SS, Elsalahaty MI, Ismail DF, Radwan MA, Elkafas SS, Loutfy SA, Elshazli RM, Baazaoui N, Ahmed AE, Hafez W, Diab M, Sakran M, El-Saadony MT, El-Tarabily KA, Kamal HK, Hessien M. The emerging roles of sphingosine 1-phosphate and SphK1 in cancer resistance: a promising therapeutic target. Cancer Cell Int 2024; 24:89. [PMID: 38419070 PMCID: PMC10903003 DOI: 10.1186/s12935-024-03221-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/09/2024] [Indexed: 03/02/2024] Open
Abstract
Cancer chemoresistance is a problematic dilemma that significantly restrains numerous cancer management protocols. It can promote cancer recurrence, spreading of cancer, and finally, mortality. Accordingly, enhancing the responsiveness of cancer cells towards chemotherapies could be a vital approach to overcoming cancer chemoresistance. Tumour cells express a high level of sphingosine kinase-1 (SphK1), which acts as a protooncogenic factor and is responsible for the synthesis of sphingosine-1 phosphate (S1P). S1P is released through a Human ATP-binding cassette (ABC) transporter to interact with other phosphosphingolipids components in the interstitial fluid in the tumor microenvironment (TME), provoking communication, progression, invasion, and tumor metastasis. Also, S1P is associated with several impacts, including anti-apoptotic behavior, metastasis, mesenchymal transition (EMT), angiogenesis, and chemotherapy resistance. Recent reports addressed high levels of S1P in several carcinomas, including ovarian, prostate, colorectal, breast, and HCC. Therefore, targeting the S1P/SphK signaling pathway is an emerging therapeutic approach to efficiently attenuate chemoresistance. In this review, we comprehensively discussed S1P functions, metabolism, transport, and signaling. Also, through a bioinformatic framework, we pointed out the alterations of SphK1 gene expression within different cancers with their impact on patient survival, and we demonstrated the protein-protein network of SphK1, elaborating its sparse roles. Furthermore, we made emphasis on different machineries of cancer resistance and the tight link with S1P. We evaluated all publicly available SphK1 inhibitors and their inhibition activity using molecular docking and how SphK1 inhibitors reduce the production of S1P and might reduce chemoresistance, an approach that might be vital in the course of cancer treatment and prognosis.
Collapse
Affiliation(s)
- Samar Sami Alkafaas
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Mohamed I Elsalahaty
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doha F Ismail
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mustafa Ali Radwan
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sara Samy Elkafas
- Production Engineering and Mechanical Design Department, Faculty of Engineering, Menofia University, Menofia, Egypt
- Faculty of Control System and Robotics, ITMO University, Saint-Petersburg, 197101, Russia
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
- Nanotechnology Research Center, British University, Cairo, Egypt
| | - Rami M Elshazli
- Biochemistry and Molecular Genetics Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University-Egypt, New Damietta, 34517, Egypt
| | - Narjes Baazaoui
- Biology Department, College of Sciences and Arts Muhayil Assir, King Khalid University, Abha 61421, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha 61413, Saudi Arabia
| | - Wael Hafez
- NMC Royal Hospital, 16th Street, 35233, Khalifa, Abu Dhabi, United Arab Emirates
- Medical Research Division, Department of Internal Medicine, The National Research Centre, Cairo 11511, Egypt
| | - Mohanad Diab
- Burjeel Hospital Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Mohamed Sakran
- Biochemistry Division, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig 44511, Egypt
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| | - Hani K Kamal
- Anatomy and Histology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed Hessien
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
11
|
Casari M, Siegl D, Deppermann C, Schuppan D. Macrophages and platelets in liver fibrosis and hepatocellular carcinoma. Front Immunol 2023; 14:1277808. [PMID: 38116017 PMCID: PMC10728659 DOI: 10.3389/fimmu.2023.1277808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
During fibrosis, (myo)fibroblasts deposit large amounts of extracellular matrix proteins, thereby replacing healthy functional tissue. In liver fibrosis, this leads to the loss of hepatocyte function, portal hypertension, variceal bleeding, and increased susceptibility to infection. At an early stage, liver fibrosis is a dynamic and reversible process, however, from the cirrhotic stage, there is significant progression to hepatocellular carcinoma. Both liver-resident macrophages (Kupffer cells) and monocyte-derived macrophages are important drivers of fibrosis progression, but can also induce its regression once triggers of chronic inflammation are eliminated. In liver cancer, they are attracted to the tumor site to become tumor-associated macrophages (TAMs) polarized towards a M2- anti-inflammatory/tumor-promoting phenotype. Besides their role in thrombosis and hemostasis, platelets can also stimulate fibrosis and tumor development by secreting profibrogenic factors and regulating the innate immune response, e.g., by interacting with monocytes and macrophages. Here, we review recent literature on the role of macrophages and platelets and their interplay in liver fibrosis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Martina Casari
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Dominik Siegl
- Institute for Translational Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Carsten Deppermann
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immune Therapy Forschungszentrum für Immuntherapie (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Detlef Schuppan
- Institute for Translational Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immune Therapy Forschungszentrum für Immuntherapie (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Hammad SM, Lopes-Virella MF. Circulating Sphingolipids in Insulin Resistance, Diabetes and Associated Complications. Int J Mol Sci 2023; 24:14015. [PMID: 37762318 PMCID: PMC10531201 DOI: 10.3390/ijms241814015] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Sphingolipids play an important role in the development of diabetes, both type 1 and type 2 diabetes, as well as in the development of both micro- and macro-vascular complications. Several reviews have been published concerning the role of sphingolipids in diabetes but most of the emphasis has been on the possible mechanisms by which sphingolipids, mainly ceramides, contribute to the development of diabetes. Research on circulating levels of the different classes of sphingolipids in serum and in lipoproteins and their importance as biomarkers to predict not only the development of diabetes but also of its complications has only recently emerged and it is still in its infancy. This review summarizes the previously published literature concerning sphingolipid-mediated mechanisms involved in the development of diabetes and its complications, focusing on how circulating plasma sphingolipid levels and the relative content carried by the different lipoproteins may impact their role as possible biomarkers both in the development of diabetes and mainly in the development of diabetic complications. Further studies in this field may open new therapeutic avenues to prevent or arrest/reduce both the development of diabetes and progression of its complications.
Collapse
Affiliation(s)
- Samar M. Hammad
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Maria F. Lopes-Virella
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson VA Medical Center, Charleston, SC 29425, USA
| |
Collapse
|
13
|
Abdelraheem KM, Younis NN, Shaheen MA, Elswefy SE, Ali SI. Raspberry ketone improves non-alcoholic fatty liver disease induced in rats by modulating sphingosine kinase/sphingosine-1-phosphate and toll-like receptor 4 pathways. J Pharm Pharmacol 2023:7160323. [PMID: 37167472 DOI: 10.1093/jpp/rgad044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 04/19/2023] [Indexed: 05/13/2023]
Abstract
OBJECTIVES To investigate the therapeutic role of calorie-restricted diet (CR) and raspberry ketone (RK) in non-alcoholic fatty liver disease (NAFLD) and the implication of sphingosine kinase-1 (SphK1)/sphingosine-1-phosphate (S1P) and toll-like receptor 4 (TLR4) signalling. METHODS NAFLD was induced by feeding rats high-fat-fructose-diet (HFFD) for 6 weeks. Rats were then randomly assigned to three groups (n = 6 each); NAFLD group continued on HFFD for another 8 weeks. CR group was switched to CR diet (25% calorie restriction) for 8 weeks and RK group was switched to normal diet and received RK (55 mg/kg/day; orally) for 8 weeks. Another six rats were used as normal control. KEY FINDINGS HFFD induced a state of NAFLD indicated by increased fat deposition in liver tissue along with dyslipidemia, elevated liver enzymes, oxidative stress and inflammation. Either CR diet or RK reversed these changes and decreased HFFD-induced elevation of hepatic SphK1, S1P, S1PR1 and TLR4. Of notice, RK along with a normal calorie diet was even better than CR alone in most studied parameters. CONCLUSIONS SphK1/S1P and TLR4 are interconnected and related to the establishment of HFFD-induced NAFLD and can be modulated by RK. Supplementation of RK without calorie restriction to patients with NAFLD unable to follow CR diet to achieve their treatment goals would be a promising therapeutic modality.
Collapse
Affiliation(s)
- Kareem M Abdelraheem
- Biochemistry Department, Faculty of Pharmacy, Sinai University - Qantara Branch, Ismailia, Egypt
| | - Nahla N Younis
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed A Shaheen
- Histology and Cell Biology Department, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Sahar E Elswefy
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
- Biochemistry Department, Faculty of Pharmacy, Delta University for Sciences and Technology, Gamasa, Egypt
| | - Sousou I Ali
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
14
|
Alessenko AV, Gutner UA, Shupik MA. Involvement of Lipids in the Pathogenesis of Amyotrophic Lateral Sclerosis. Life (Basel) 2023; 13:life13020510. [PMID: 36836867 PMCID: PMC9966871 DOI: 10.3390/life13020510] [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: 12/06/2022] [Revised: 01/26/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive degeneration of upper and lower motor neurons. To study its underlying mechanisms, a variety of models are currently used at the cellular level and in animals with mutations in multiple ALS associated genes, including SOD1, C9ORF72, TDP-43, and FUS. Key mechanisms involved in the disease include excitotoxicity, oxidative stress, mitochondrial dysfunction, neuroinflammatory, and immune reactions. In addition, significant metabolism alterations of various lipids classes, including phospholipids, fatty acids, sphingolipids, and others have been increasingly recognized. Recently, the mechanisms of programmed cell death (apoptosis), which may be responsible for the degeneration of motor neurons observed in the disease, have been intensively studied. In this context, sphingolipids, which are the most important sources of secondary messengers transmitting signals for cell proliferation, differentiation, and apoptosis, are gaining increasing attention in the context of ALS pathogenesis given their role in the development of neuroinflammatory and immune responses. This review describes changes in lipids content and activity of enzymes involved in their metabolism in ALS, both summarizing current evidence from animal models and clinical studies and discussing the potential of new drugs among modulators of lipid metabolism enzymes.
Collapse
|
15
|
Chen H, Haddadi N, Zhu X, Hatoum D, Chen S, Nassif NT, Lin Y, McGowan EM. Expression Profile of Sphingosine Kinase 1 Isoforms in Human Cancer Tissues and Cells: Importance and Clinical Relevance of the Neglected 1b-Isoform. JOURNAL OF ONCOLOGY 2022; 2022:2250407. [PMID: 36532885 PMCID: PMC9750787 DOI: 10.1155/2022/2250407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/12/2022] [Accepted: 11/17/2022] [Indexed: 09/28/2023]
Abstract
Background Overexpression of sphingosine kinase 1 (SphK1) is casually associated with many types of cancer, and inhibitors of SphK1 sensitize tumors to chemotherapy. SphK1 is expressed as two major isoforms, SphK1a and SphK1b. To date, no information has been reported on the SphK1 isoform expression profile and its clinical relevance. Objective The objective is to examine the expression profile of the SphK1a and SPhK1b isoforms in human cancer and noncancer tissues and cell lines and explore their clinical relevance. Methods We used PCR to qualitatively examine the expression profile of these two isoforms in breast, liver, and prostate cancer tissues plus paired adjacent tissues and in 11 cancer and normal cell lines (breast, cervical, bone, prostate, colon, brain, mesothelioma tumor and benign, and human kidney cells). Results We found that SphK1a was ubiquitously expressed in all cancer cells and tissues tested; in contrast, SphK1b was only expressed in selective cell types in breast, prostate, and lung cancer. Conclusions Our data suggest that SphK1a is important for generic SphK1/S1P functions, and SphK1b mediates specialized and/or unique pathways in a specific type of tissue and could be a biomarker for cancer. This discovery is important for future SphK1-related cancer research and may have clinical implications in drug development associated with SphK1-directed cancer treatment.
Collapse
Affiliation(s)
- Hongjie Chen
- Department of Traditional Chinese Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Nahal Haddadi
- School of Life Sciences, University of Technology Sydney, Broadway, NSW, Australia
| | - Xiaofeng Zhu
- Department of Transplant Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Diana Hatoum
- School of Life Sciences, University of Technology Sydney, Broadway, NSW, Australia
- Public Health and College of Arts and Sciences, Phoenicia University, Daoudiye, Lebanon
| | - Size Chen
- Central Laboratory, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Najah T. Nassif
- School of Life Sciences, University of Technology Sydney, Broadway, NSW, Australia
| | - Yiguang Lin
- Department of Traditional Chinese Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- School of Life Sciences, University of Technology Sydney, Broadway, NSW, Australia
- Central Laboratory, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Eileen M. McGowan
- School of Life Sciences, University of Technology Sydney, Broadway, NSW, Australia
- Central Laboratory, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
16
|
Paranjpe V, Galor A, Grambergs R, Mandal N. The role of sphingolipids in meibomian gland dysfunction and ocular surface inflammation. Ocul Surf 2022; 26:100-110. [PMID: 35973562 PMCID: PMC10259413 DOI: 10.1016/j.jtos.2022.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 11/26/2022]
Abstract
Inflammation occurs in response to tissue injury and invasion of microorganisms and is carried out by the innate and adaptive immune systems, which are regulated by numerous chemokines, cytokines, and lipid mediators. There are four major families of bioactive lipid mediators that play an integral role in inflammation - eicosanoids, sphingolipids (SPL), specialized pro-resolving mediators (SPM), and endocannabinoids. SPL have been historically recognized as important structural components of cellular membranes; their roles as bioactive lipids and inflammatory mediators are recent additions. Major SPL metabolites, including sphingomyelin, ceramide, ceramide 1-phosphate (C1P), sphingosine, sphingosine 1-phosphate (S1P), and their respective enzymes have been studied extensively, primarily in cell-culture and animal models, for their roles in cellular signaling and regulating inflammation and apoptosis. Less focus has been given to the involvement of SPL in eye diseases. As such, the aim of this review was to examine relationships between the SPL family and ocular surface diseases, focusing on their role in disease pathophysiology and discussing the potential of therapeutics that disrupt SPL pathways.
Collapse
Affiliation(s)
- Vikram Paranjpe
- Department of Ophthalmology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA
| | - Anat Galor
- Miami Veterans Administration Medical Center, 1201 NW 16th St, Miami, FL, 33125, USA; Bascom Palmer Eye Institute, University of Miami, 900 NW 17th Street, Miami, FL, 33136, USA.
| | - Richard Grambergs
- Departments of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Sciences Center, Hamilton Eye Institute, 930 Madison Avenue, Memphis, TN, 38163, USA
| | - Nawajes Mandal
- Departments of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Sciences Center, Hamilton Eye Institute, 930 Madison Avenue, Memphis, TN, 38163, USA; Memphis VA Medical Center, Memphis, TN, 38104, USA.
| |
Collapse
|
17
|
Kołakowski A, Dziemitko S, Chmielecka A, Żywno H, Bzdęga W, Charytoniuk T, Chabowski A, Konstantynowicz-Nowicka K. Molecular Advances in MAFLD—A Link between Sphingolipids and Extracellular Matrix in Development and Progression to Fibrosis. Int J Mol Sci 2022; 23:ijms231911380. [PMID: 36232681 PMCID: PMC9569877 DOI: 10.3390/ijms231911380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/18/2022] [Accepted: 09/23/2022] [Indexed: 11/25/2022] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is a major cause of liver diseases globally and its prevalence is expected to grow in the coming decades. The main cause of MAFLD development is changed in the composition of the extracellular matrix (ECM). Increased production of matrix molecules and inflammatory processes lead to progressive fibrosis, cirrhosis, and ultimately liver failure. In addition, increased accumulation of sphingolipids accompanied by increased expression of pro-inflammatory cytokines in the ECM is closely related to lipogenesis, MAFLD development, and its progression to fibrosis. In our work, we will summarize all information regarding the role of sphingolipids e.g., ceramide and S1P in MAFLD development. These sphingolipids seem to have the most significant effect on macrophages and, consequently, HSCs which trigger the entire cascade of overproduction matrix molecules, especially type I and III collagen, proteoglycans, elastin, and also tissue inhibitors of metalloproteinases, which as a result cause the development of liver fibrosis.
Collapse
Affiliation(s)
- Adrian Kołakowski
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Sylwia Dziemitko
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | | | - Hubert Żywno
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Wiktor Bzdęga
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Tomasz Charytoniuk
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
- Department of Ophthalmology, Antoni Jurasz University Hospital No. 1, 85-094 Bydgoszcz, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland
| | | |
Collapse
|
18
|
Varre JV, Holland WL, Summers SA. You aren't IMMUNE to the ceramides that accumulate in cardiometabolic disease. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159125. [PMID: 35218934 PMCID: PMC9050903 DOI: 10.1016/j.bbalip.2022.159125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 02/14/2022] [Indexed: 02/06/2023]
Abstract
Obesity leads to persistent increases in immune responses that contribute to cardiometabolic pathologies such as diabetes and cardiovascular disease. Pro-inflammatory macrophages infiltrate the expanding fat mass, which leads to increased production of cytokines such as tumor necrosis factor-alpha. Moreover, saturated fatty acids enhance signaling through the toll-like receptors involved in innate immunity. Herein we discuss the evidence that ceramides-which are intermediates in the biosynthetic pathway that produces sphingolipids-are essential intermediates that link these inflammatory signals to impaired tissue function. We discuss the mechanisms linking these immune insults to ceramide production and review the numerous ceramide actions that alter cellular metabolism, induce oxidative stress, and stimulate apoptosis. Lastly, we evaluate the correlation of ceramides in humans with inflammation-linked cardiometabolic disease and discuss preclinical studies which suggest that ceramide-lowering interventions may be an effective strategy to treat or prevent such maladies.
Collapse
Affiliation(s)
- Joseph V Varre
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 94108, United States of America
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 94108, United States of America
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 94108, United States of America.
| |
Collapse
|
19
|
Biomarkers for the Detection and Management of Hepatocellular Carcinoma in Patients Treated with Direct-Acting Antivirals. Cancers (Basel) 2022; 14:cancers14112700. [PMID: 35681679 PMCID: PMC9179595 DOI: 10.3390/cancers14112700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Chronic Hepatitis C virus (HCV) represents the main etiological factor for hepatocellular carcinoma (HCC) in developed countries. The introduction of direct-acting antivirals (DAAs) improved the eradication of the hepatitis C virus (HCV) but not the reduction in the incidence of HCV-associated HCC. Some patients still develop HCC, even after reaching a sustained virological response (SVR). This review is a summary of pre-clinical studies that investigated predictive biomarkers for HCC occurrence and recurrence in HCV-infected patients treated with DAAs. The presented biomarkers are found dysregulated in serum or tissue at specific time points (before, during, after DAA treatment or post SVR) and correlated with HCC-predisposing conditions. Thus, this review aims to improve the management of patients developing HCV-induced HCC. Abstract Hepatocellular carcinoma (HCC) is the sixth-most common type of cancer worldwide and chronic Hepatitis C virus (HCV) represents the main etiological factor in developed countries. HCV promotes hepatocarcinogenesis through persistent liver inflammation and dysregulation of cell signaling pathways. The introduction of direct-acting antivirals (DAAs) resulted in a significant improvement in the eradication of the virus, with an expected reduction of HCC incidence. However, the risk of HCC development can persist after DAA treatment. Recent studies have investigated the potential use of molecular biomarkers that predict HCC occurrence or recurrence helping the stratification of patients under surveillance. This review aimed to summarize all pre-clinical exploration of predictive biomarkers to identify DAA-treated patients at risk for HCC development. Dysregulated microRNAs, lncRNAs, histone modifications, cytokines, proteins, and sphingolipids represent various classes of HCC risk predictors identified in two different biological sources (tissue and serum). The non-invasive serum markers can provide a more accessible means to perform clinical monitoring and predict the risk of HCC. In addition, conditions like cirrhosis, predisposing to HCC, strongly correlate with most of the molecular predictors identified, supporting the value of these molecules as possible biomarkers of HCC in DAA-treated patients.
Collapse
|
20
|
Jackson KG, Way GW, Zhou H. Bile acids and sphingolipids in non-alcoholic fatty liver disease. Chin Med J (Engl) 2022; 135:1163-1171. [PMID: 35788089 PMCID: PMC9337250 DOI: 10.1097/cm9.0000000000002156] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Indexed: 12/14/2022] Open
Abstract
ABSTRACT Non-alcoholic fatty liver disease (NAFLD) is one of the fastest-growing diseases, and its global prevalence is estimated to increase >50% by 2030. NAFLD is comorbid with metabolic syndrome, obesity, type 2 diabetes, and insulin resistance. Despite extensive research efforts, there are no pharmacologic or biological therapeutics for the treatment of NAFLD. Bile acids and sphingolipids are well-characterized signaling molecules. Over the last few decades, researchers have uncovered potential mechanisms by which bile acids and sphingolipids regulate hepatic lipid metabolism. Dysregulation of bile acid and sphingolipid metabolism has been linked to steatosis, inflammation, and fibrosis in patients with NAFLD. This clinical observation has been recapitulated in animal models, which are well-accepted by experts in the hepatology field. Recent transcriptomic and lipidomic studies also show that sphingolipids are important players in the pathogenesis of NAFLD. Moreover, the identification of bile acids as activators of sphingolipid-mediated signaling pathways established a novel theory for bile acid and sphingolipid biology. In this review, we summarize the recent advances in the understanding of bile acid and sphingolipid-mediated signaling pathways as potential contributors to NAFLD. A better understanding of the pathologic effects mediated by bile acids and sphingolipids will facilitate the development of new diagnostic and therapeutic strategies for NAFLD.
Collapse
Affiliation(s)
- Kaitlyn G. Jackson
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Grayson W. Way
- Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
- Central Virginia Veterans Healthcare System, Richmond, VA 23249, USA
| |
Collapse
|
21
|
Dalbeni A, Castelli M, Zoncapè M, Minuz P, Sacerdoti D. Platelets in Non-alcoholic Fatty Liver Disease. Front Pharmacol 2022; 13:842636. [PMID: 35250588 PMCID: PMC8895200 DOI: 10.3389/fphar.2022.842636] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/03/2022] [Indexed: 12/17/2022] Open
Abstract
Non alcoholic steatohepatitis (NASH) is the inflammatory reaction of the liver to excessive accumulation of lipids in the hepatocytes. NASH can progress to cirrhosis and hepatocellular carcinoma (HCC). Fatty liver is the hepatic manifestation of metabolic syndrome. A subclinical inflammatory state is present in patients with metabolic alterations like insulin resistance, type-2 diabetes, obesity, hyperlipidemia, and hypertension. Platelets participate in immune cells recruitment and cytokines-induced liver damage. It is hypothesized that lipid toxicity cause accumulation of platelets in the liver, platelet adhesion and activation, which primes the immunoinflammatory reaction and activation of stellate cells. Recent data suggest that antiplatelet drugs may interrupt this cascade and prevent/improve NASH. They may also improve some metabolic alterations. The pathophysiology of inflammatory liver disease and the implication of platelets are discussed in details.
Collapse
Affiliation(s)
- Andrea Dalbeni
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Marco Castelli
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Mirko Zoncapè
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Pietro Minuz
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- *Correspondence: Pietro Minuz,
| | - David Sacerdoti
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| |
Collapse
|
22
|
McGowan EM, Lin Y, Chen S. Targeting Chronic Inflammation of the Digestive System in Cancer Prevention: Modulators of the Bioactive Sphingolipid Sphingosine-1-Phosphate Pathway. Cancers (Basel) 2022; 14:cancers14030535. [PMID: 35158806 PMCID: PMC8833440 DOI: 10.3390/cancers14030535] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 01/04/2023] Open
Abstract
Incidence of gastrointestinal (GI) cancers is increasing, and late-stage diagnosis makes these cancers difficult to treat. Chronic and low-grade inflammation are recognized risks for most GI cancers. The GI mucosal immune system maintains healthy homeostasis and signalling molecules made from saturated fats, bioactive sphingolipids, play essential roles in healthy GI immunity. Sphingosine-1-phosphate (S1P), a bioactive sphingolipid, is a key mediator in a balanced GI immune response. Disruption in the S1P pathway underlies systemic chronic metabolic inflammatory disorders, including diabetes and GI cancers, providing a strong rationale for using modulators of the S1P pathway to treat pathological inflammation. Here, we discuss the effects of bioactive sphingolipids in immune homeostasis with a focus on S1P in chronic low-grade inflammation associated with increased risk of GI carcinogenesis. Contemporary information on S1P signalling involvement in cancers of the digestive system, from top to bottom, is reviewed. Further, we discuss the use of novel S1P receptor modulators currently in clinical trials and their potential as first-line drugs in the clinic for chronic inflammatory diseases. Recently, ozanimod (ZeposiaTM) and etrasimod have been approved for clinical use to treat ulcerative colitis and eosinophilic oesophagitis, respectively, which may have longer term benefits in reducing risk of GI cancers.
Collapse
Affiliation(s)
- Eileen M. McGowan
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China; (Y.L.); (S.C.)
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precise Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia
- Correspondence: ; Tel.: +86-614-0581-4048
| | - Yiguang Lin
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China; (Y.L.); (S.C.)
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precise Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
- School of Life Sciences, University of Technology Sydney, Broadway, Sydney, NSW 2007, Australia
| | - Size Chen
- Central Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China; (Y.L.); (S.C.)
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precise Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| |
Collapse
|
23
|
Gutner UA, Shupik MA. The Role of Sphingosine-1-Phosphate in Neurodegenerative Diseases. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2021. [DOI: 10.1134/s1068162021050277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Hong CH, Ko MS, Kim JH, Cho H, Lee CH, Yoon JE, Yun JY, Baek IJ, Jang JE, Lee SE, Cho YK, Baek JY, Oh SJ, Lee BY, Lim JS, Lee J, Hartig SM, Conde de la Rosa L, Garcia-Ruiz C, Lee KU, Fernández-Checa JC, Choi JW, Kim S, Koh EH. Sphingosine 1-Phosphate Receptor 4 Promotes Nonalcoholic Steatohepatitis by Activating NLRP3 Inflammasome. Cell Mol Gastroenterol Hepatol 2021; 13:925-947. [PMID: 34890841 PMCID: PMC8810559 DOI: 10.1016/j.jcmgh.2021.12.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Sphingosine 1-phosphate receptors (S1PRs) are a group of G-protein-coupled receptors that confer a broad range of functional effects in chronic inflammatory and metabolic diseases. S1PRs also may mediate the development of nonalcoholic steatohepatitis (NASH), but the specific subtypes involved and the mechanism of action are unclear. METHODS We investigated which type of S1PR isoforms is activated in various murine models of NASH. The mechanism of action of S1PR4 was examined in hepatic macrophages isolated from high-fat, high-cholesterol diet (HFHCD)-fed mice. We developed a selective S1PR4 functional antagonist by screening the fingolimod (2-amino-2-[2-(4- n -octylphenyl)ethyl]-1,3- propanediol hydrochloride)-like sphingolipid-focused library. RESULTS The livers of various mouse models of NASH as well as hepatic macrophages showed high expression of S1pr4. Moreover, in a cohort of NASH patients, expression of S1PR4 was 6-fold higher than those of healthy controls. S1pr4+/- mice were protected from HFHCD-induced NASH and hepatic fibrosis without changes in steatosis. S1pr4 depletion in hepatic macrophages inhibited lipopolysaccharide-mediated Ca++ release and deactivated the Nod-like receptor pyrin domain-containning protein 3 (NLRP3) inflammasome. S1P increased the expression of S1pr4 in hepatic macrophages and activated NLRP3 inflammasome through inositol trisphosphate/inositol trisphosphate-receptor-dependent [Ca++] signaling. To further clarify the biological function of S1PR4, we developed SLB736, a novel selective functional antagonist of SIPR4. Similar to S1pr4+/- mice, administration of SLB736 to HFHCD-fed mice prevented the development of NASH and hepatic fibrosis, but not steatosis, by deactivating the NLRP3 inflammasome. CONCLUSIONS S1PR4 may be a new therapeutic target for NASH that mediates the activation of NLRP3 inflammasome in hepatic macrophages.
Collapse
Affiliation(s)
- Chung Hwan Hong
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Myoung Seok Ko
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Hyun Kim
- College of Pharmacy, Seoul National University, Seoul, Korea,College of Pharmacy, Kangwon National University, Chuncheon, Korea
| | - Hyunkyung Cho
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Chi-Ho Lee
- College of Pharmacy, Gachon University, Incheon, Korea
| | - Ji Eun Yoon
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji-Young Yun
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - In-Jeoung Baek
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jung Eun Jang
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung Eun Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yun Kyung Cho
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ji Yeon Baek
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo Jin Oh
- New Drug Development Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | - Joon Seo Lim
- Clinical Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jongkook Lee
- College of Pharmacy, Kangwon National University, Chuncheon, Korea
| | - Sean M. Hartig
- Molecular and Cellular Biology, Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, Texas
| | - Laura Conde de la Rosa
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona and Liver Unit-Hospital Clinic-Instituto de Investigaciones Biomédicas August Pi i Sunyer, Centro de Investigación Biomédica en Red, Barcelona, Spain
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona and Liver Unit-Hospital Clinic-Instituto de Investigaciones Biomédicas August Pi i Sunyer, Centro de Investigación Biomédica en Red, Barcelona, Spain,Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ki-Up Lee
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jose C. Fernández-Checa
- Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona and Liver Unit-Hospital Clinic-Instituto de Investigaciones Biomédicas August Pi i Sunyer, Centro de Investigación Biomédica en Red, Barcelona, Spain,Research Center for Alcoholic Liver and Pancreatic Diseases and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, California,Correspondence Address correspondence to: Jose C. Fernández-Checa, PhD, Department of Cell Death and Proliferation, Instituto Investigaciones Biomédicas de Barcelona, Consejo Superior de Investigaciones Cientificas, Barcelona and Liver Unit-Hospital Clinic–Instituto de Investigaciones Biomédicas August Pi i Sunyer, Centro de Investigación Biomédica en Red, Barcelona 08036, Spain. fax: (34) 93-3129405.
| | - Ji Woong Choi
- College of Pharmacy, Gachon University, Incheon, Korea,Ji Woong Choi, PhD, Laboratory of Pharmacology, College of Pharmacy, Gachon University, 191 Hambakmoero, Yeonsu-gu, Incheon 21936, Korea. fax: (82) 32-820-4829.
| | - Sanghee Kim
- College of Pharmacy, Seoul National University, Seoul, Korea,Sanghee Kim, PhD, College of Pharmacy, Seoul National University, Gwanak-ro, Gwanak-gu, Seoul 08826, Korea. fax: (82) 2-762-8322.
| | - Eun Hee Koh
- Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea,Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea,Eun Hee Koh, MD, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea. fax: (82) 2-3010-6962.
| |
Collapse
|
25
|
Alessenko AV, Gutner UA, Nebogatikov VO, Shupik MA, Ustyugov AA. [The role of sphingolipids in pathogenesis of amyotrophic lateral sclerosis]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:131-140. [PMID: 34481449 DOI: 10.17116/jnevro2021121081131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disease characterized by selective degeneration of motor neurons of the spinal cord and motor cortex and brain stem. The key features of the course of this disease are excitotoxicity, oxidative stress, mitochondrial dysfunction, neuro-inflammatory and immune reactions. Recently, the mechanisms of programmed cell death (apoptosis), which may be responsible for the degeneration of motor neurons in this disease, have been intensively studied. In this regard, sphingolipids, which are the most important sources of secondary messengers that transmit cell proliferation, differentiation and apoptosis signals, and are involved in the development of neuroinflammatory and immune responses, are of particular interest in the context of ALS pathogenesis. The review provides information from domestic and foreign authors on the involvement of various sphingolipids (sphingomyelins, ceramides, sphingosine, sphinganin, sphingosine-1-phosphate, galactosylceramides, glucosylceramides, gangliosides) in the development of pro-inflammatory reactions and apoptosis of motor neurons in ALS. The authors discuss the prospects of using new drugs that control the metabolism of sphingolipids for the treatment of ALS.
Collapse
Affiliation(s)
| | - U A Gutner
- Institute of Biochemical Physic, Moscow, Russia
| | - V O Nebogatikov
- Institute of Physiologically Active Compounds, Chernogolovka, Russia
| | - M A Shupik
- Institute of Biochemical Physic, Moscow, Russia
| | - A A Ustyugov
- Institute of Physiologically Active Compounds, Chernogolovka, Russia
| |
Collapse
|
26
|
Green CD, Maceyka M, Cowart LA, Spiegel S. Sphingolipids in metabolic disease: The good, the bad, and the unknown. Cell Metab 2021; 33:1293-1306. [PMID: 34233172 PMCID: PMC8269961 DOI: 10.1016/j.cmet.2021.06.006] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/05/2021] [Accepted: 06/11/2021] [Indexed: 01/10/2023]
Abstract
The bioactive sphingolipid metabolites ceramide and sphingosine-1-phosphate (S1P) are a recent addition to the lipids accumulated in obesity and have emerged as important molecular players in metabolic diseases. Here we summarize evidence that dysregulation of sphingolipid metabolism correlates with pathogenesis of metabolic diseases in humans. This review discusses the current understanding of how ceramide regulates signaling and metabolic pathways to exacerbate metabolic diseases and the Janus faces for its further metabolite S1P, the kinases that produce it, and the multifaceted and at times opposing actions of S1P receptors in various tissues. Gaps and limitations in current knowledge are highlighted together with the need to further decipher the full array of their actions in tissue dysfunction underlying metabolic pathologies, pointing out prospects to move this young field of research toward the development of effective therapeutics.
Collapse
Affiliation(s)
- Christopher D Green
- Department of Biochemistry and Molecular Biology, VCU School of Medicine and Massey Cancer Center, Richmond, VA 23298, USA
| | - Michael Maceyka
- Department of Biochemistry and Molecular Biology, VCU School of Medicine and Massey Cancer Center, Richmond, VA 23298, USA
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology, VCU School of Medicine and Massey Cancer Center, Richmond, VA 23298, USA; Hunter Holmes McGuire VA Medical Center, Richmond, VA 23298, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, VCU School of Medicine and Massey Cancer Center, Richmond, VA 23298, USA.
| |
Collapse
|
27
|
Abed Rabbo M, Khodour Y, Kaguni LS, Stiban J. Sphingolipid lysosomal storage diseases: from bench to bedside. Lipids Health Dis 2021; 20:44. [PMID: 33941173 PMCID: PMC8094529 DOI: 10.1186/s12944-021-01466-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/14/2021] [Indexed: 01/13/2023] Open
Abstract
Johann Ludwig Wilhelm Thudicum described sphingolipids (SLs) in the late nineteenth century, but it was only in the past fifty years that SL research surged in importance and applicability. Currently, sphingolipids and their metabolism are hotly debated topics in various biochemical fields. Similar to other macromolecular reactions, SL metabolism has important implications in health and disease in most cells. A plethora of SL-related genetic ailments has been described. Defects in SL catabolism can cause the accumulation of SLs, leading to many types of lysosomal storage diseases (LSDs) collectively called sphingolipidoses. These diseases mainly impact the neuronal and immune systems, but other systems can be affected as well. This review aims to present a comprehensive, up-to-date picture of the rapidly growing field of sphingolipid LSDs, their etiology, pathology, and potential therapeutic strategies. We first describe LSDs biochemically and briefly discuss their catabolism, followed by general aspects of the major diseases such as Gaucher, Krabbe, Fabry, and Farber among others. We conclude with an overview of the available and potential future therapies for many of the diseases. We strive to present the most important and recent findings from basic research and clinical applications, and to provide a valuable source for understanding these disorders.
Collapse
Affiliation(s)
- Muna Abed Rabbo
- Department of Biology and Biochemistry, Birzeit University, P.O. Box 14, Ramallah, West Bank, 627, Palestine
| | - Yara Khodour
- Department of Biology and Biochemistry, Birzeit University, P.O. Box 14, Ramallah, West Bank, 627, Palestine
| | - Laurie S Kaguni
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Johnny Stiban
- Department of Biology and Biochemistry, Birzeit University, P.O. Box 14, Ramallah, West Bank, 627, Palestine.
| |
Collapse
|
28
|
Squecco R, Pierucci F, Idrizaj E, Frati A, Lenci E, Vicenti C, Iachini MC, Martinesi M, Garella R, Baccari MC, Francini F, Meacci E. Ceramide/protein phosphatase 2A axis is engaged in gap junction impairment elicited by PCB153 in liver stem-like progenitor cells. Mol Cell Biochem 2021; 476:3111-3126. [PMID: 33837873 PMCID: PMC8263450 DOI: 10.1007/s11010-021-04135-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 03/11/2021] [Indexed: 12/22/2022]
Abstract
The widespread environmental pollutant 2,2',4,4',5,5'-hexachlorobiphenyl (PCB153) is a non-dioxin-like toxicant. It is a potential carcinogen compound able to induce gap junction (GJ) intercellular communication impairment, probably the first non-genomic event leading to tumor promotion. Although PCBs have been known for many years, the molecular mode of PCB153 action is still unclear. Recent studies from our research group have shown that the toxicant elicits a transient modulation of connexin (Cx) 43-formed GJs in hepatic stem-like WB-F344 cells involving sphingosine 1-phosphate (S1P) path. Taking into account that other strictly related bioactive sphingolipids, such as ceramide (Cer), may have different effects from S1P, here we aim to clarify the signaling paths engaged by PCB153 in the control of GJs, focusing primarily on the role of Cer. Accordingly, we have achieved a combined biomolecular and electrophysiological analysis of GJs in cultured WB-F344 cells treated with PCB153 at different time points. We have found that the toxicant elicited a time-dependent regulation of GJs formed by different Cx isoforms, through a transient modulation of Cer/Cer kinase (CerK) axis and, in turn, of protein phosphatase 2A (PP2A). Our new findings demonstrate the existence of a specific molecular mechanism downstream to Cer, which distinctly affects the voltage-dependent and -independent GJs in liver stem-like cells, and open new opportunities for the identification of additional potential targets of these environmental toxicants.
Collapse
Affiliation(s)
- Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Viale GB Morgagni 63, 50134, Florence, Italy
| | - Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Research unit of Molecular and Applied Biology, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Viale GB Morgagni 63, 50134, Florence, Italy
| | - Alessia Frati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Research unit of Molecular and Applied Biology, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Elena Lenci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Research unit of Molecular and Applied Biology, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Catia Vicenti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Research unit of Molecular and Applied Biology, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Maria Chiara Iachini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Research unit of Molecular and Applied Biology, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Maria Martinesi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Research unit of Molecular and Applied Biology, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Rachele Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Viale GB Morgagni 63, 50134, Florence, Italy
| | - Maria Caterina Baccari
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Viale GB Morgagni 63, 50134, Florence, Italy
| | - Fabio Francini
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Viale GB Morgagni 63, 50134, Florence, Italy
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Research unit of Molecular and Applied Biology, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy.
| |
Collapse
|
29
|
Lipid profile disturbances may predispose psoriatic patients to liver dysfunction. Postepy Dermatol Alergol 2021; 38:310-318. [PMID: 34408599 PMCID: PMC8362751 DOI: 10.5114/ada.2021.106209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 11/28/2019] [Indexed: 01/31/2023] Open
Abstract
INTRODUCTION Psoriasis is a chronic inflammatory disease associated with metabolic disturbances and liver dysfunction. Both serum fatty acids (FA) and ceramides (Cer) have structural functions but also are signal molecules that could be involved in the pathogenesis of liver dysfunction. AIM To assess the concentration of the circulating FA and Cer in correlation with the alanine aminotransferase (ALT) blood level in psoriatic patients. In addition, we have examined the relationship between ALT concentration and severity of the disease and inflammation markers. MATERIAL AND METHODS Eighty-five patients with psoriasis and 32 healthy controls were enrolled in the study. Patients were divided into 2 groups according to ALT blood levels. Serum concentration of 14 FA and 14 Cer were measured by gas-liquid chromatography. The results were correlated with the Psoriasis Area and Severity Index (PASI), serum lipid profile, and inflammatory markers. RESULTS We observed higher PASI score (p = 0.01) and higher C-reactive protein (p = 0.02) concentration in the group of psoriatic patients with high ALT. Serum ALT positively correlated with saturated fatty acids (SFA) (p = 0.01, r = 0.27) and SFA/unsaturated fatty acids (UFA) ratio (p = 0.01, r = 0.26). ALT negatively correlated with UFA level (p = 0.008, r = -0.28). Lignoceric ceramide positively correlated with ALT level (r = 0.22; p = 0.045) in psoriatic patients. CONCLUSIONS Patients with severe psoriasis are predisposed to the development of liver dysfunction. We have demonstrated disturbances of serum fatty acid and sphingolipid profile in psoriatic patients, which may trigger liver disease.
Collapse
|
30
|
Ceramides and sphingosine-1-phosphate mediate the distinct effects of M1/M2-macrophage infusion on liver recovery after hepatectomy. Cell Death Dis 2021; 12:324. [PMID: 33771984 PMCID: PMC7998020 DOI: 10.1038/s41419-021-03616-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 02/01/2023]
Abstract
Post-hepatectomy liver dysfunction is a life-threatening morbidity that lacks efficient therapy. Bioactive lipids involved in macrophage polarization crucially regulate tissue injury and regeneration. Herein, we investigate the key bioactive lipids that mediate the cytotherapeutic potential of polarized-macrophage for post-hepatectomy liver dysfunction. Untargeted lipidomics identified elevation of ceramide (CER) metabolites as signature lipid species relevant to M1/M2 polarization in mouse bone-marrow-derived-macrophages (BMDMs). M1 BMDMs expressed a CER-generation-metabolic pattern, leading to elevation of CER; M2 BMDMs expressed a CER-breakdown-metabolic pattern, resulting in upregulation of sphingosine-1-phosphate (S1P). After infusing M1- or M2-polarized BMDMs into the mouse liver after hepatectomy, we found that M1-BMDM infusion increased M1 polarization and CER accumulation, resulting in exaggeration of hepatocyte apoptosis and liver dysfunction. Conversely, M2-BMDM infusion enhanced M2 polarization and S1P generation, leading to alleviation of liver dysfunction with improved hepatocyte proliferation. Treatment of exogenous CER and S1P or inhibition CER and S1P synthesis by siRNA targeting relevant enzymes further revealed that CER induced apoptosis while S1P promoted proliferation in post-hepatectomy primary hepatocytes. In conclusion, CER and S1P are uncovered as critical lipid mediators for M1- and M2-polarized BMDMs to promote injury and regeneration in the liver after hepatectomy, respectively. Notably, the upregulation of hepatic S1P induced by M2-BMDM infusion may have therapeutic potential for post-hepatectomy liver dysfunction.
Collapse
|
31
|
Till Death Do Us Part-The Multifaceted Role of Platelets in Liver Diseases. Int J Mol Sci 2021; 22:ijms22063113. [PMID: 33803718 PMCID: PMC8003150 DOI: 10.3390/ijms22063113] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/09/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
Platelets are tightly connected with the liver, as both their production and their clearance are mediated by the liver. Platelets, in return, participate in a variety of liver diseases, ranging from non-alcoholic fatty liver diseases, (viral) hepatitis, liver fibrosis and hepatocellular carcinoma to liver regeneration. Due to their versatile functions, which include (1) regulation of hemostasis, (2) fine-tuning of immune responses and (3) release of growth factors and cellular mediators, platelets quickly adapt to environmental changes and modulate disease development, leading to different layers of complexity. Depending on the (patho)physiological context, platelets exert both beneficial and detrimental functions. Understanding the precise mechanisms through which platelet function is regulated at different stages of liver diseases and how platelets interact with various resident and non-resident liver cells helps to draw a clear picture of platelet-related therapeutic interventions. Therefore, this review summarizes the current knowledge on platelets in acute and chronic liver diseases and aims to shed light on how the smallest cells in the circulatory system account for changes in the (patho)physiology of the second largest organ in the human body.
Collapse
|
32
|
Yang M, Zhang CY. G protein-coupled receptors as potential targets for nonalcoholic fatty liver disease treatment. World J Gastroenterol 2021; 27:677-691. [PMID: 33716447 PMCID: PMC7934005 DOI: 10.3748/wjg.v27.i8.677] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/24/2020] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a broad-spectrum disease, ranging from simple hepatic steatosis to nonalcoholic steatohepatitis, which can progress to cirrhosis and liver cancer. Abnormal hepatic lipid accumulation is the major manifestation of this disease, and lipotoxicity promotes NAFLD progression. In addition, intermediate metabolites such as succinate can stimulate the activation of hepatic stellate cells to produce extracellular matrix proteins, resulting in progression of NAFLD to fibrosis and even cirrhosis. G protein-coupled receptors (GPCRs) have been shown to play essential roles in metabolic disorders, such as NAFLD and obesity, through their function as receptors for bile acids and free fatty acids. In addition, GPCRs link gut microbiota-mediated connections in a variety of diseases, such as intestinal diseases, hepatic steatosis, diabetes, and cardiovascular diseases. The latest findings show that gut microbiota-derived acetate contributes to liver lipogenesis by converting dietary fructose into hepatic acetyl-CoA and fatty acids. GPCR agonists, including peptides and natural products like docosahexaenoic acid, have been applied to investigate their role in liver diseases. Therapies such as probiotics and GPCR agonists may be applied to modulate GPCR function to ameliorate liver metabolism syndrome. This review summarizes the current findings regarding the role of GPCRs in the development and progression of NAFLD and describes some preclinical and clinical studies of GPCR-mediated treatment. Overall, understanding GPCR-mediated signaling in liver disease may provide new therapeutic options for NAFLD.
Collapse
Affiliation(s)
- Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
| | - Chun-Ye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65212, United States
| |
Collapse
|
33
|
Li MX, Qiang J, Bao JW, Tao YF, Zhu HJ, Xu P. Growth performance, physiological parameters, and transcript levels of lipid metabolism-related genes in hybrid yellow catfish (Tachysurus fulvidraco ♀ × Pseudobagrus vachellii ♂) fed diets containing Siberian ginseng. PLoS One 2021; 16:e0246417. [PMID: 33571255 PMCID: PMC7877655 DOI: 10.1371/journal.pone.0246417] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/19/2021] [Indexed: 01/20/2023] Open
Abstract
In high-density aquaculture, fish health can suffer because of excessive feeding, which causes fatty liver disease. Siberian ginseng (Acanthopanax senticosus) has been used as a feed additive to promote animal growth, immunity, and lipid metabolism. In this study, we explored the effects of A. senticosus on the physiology of hybrid yellow catfish (Tachysurus fulvidraco ♀ × Pseudobagrus vachellii ♂). A control group and five groups fed diets containing A. senticosus (0.5, 1, 2, 4, and 8 g A. senticosus/kg feed) were established and maintained for 8 weeks. Dietary supplementation with A. senticosus at 4 g/kg promoted growth of the hybrid yellow catfish. Serum total cholesterol (TC) and triacylglycerol (TG) levels at 2 g/kg A. senticosus (TC: 1.31 mmol/L; TG: 1.08 mmol/L) were significantly lower than in the control group (TC: 1.51 mmol/L; TG: 1.41 mmol/L), and 4 g/kg A. senticosus (17.20 μmol/g tissue) reduced the liver TG level compared with the control group (21.36 μmol/g tissue) (P <0.05). Comparative transcriptomic analysis of liver tissue between the control group and the group showing optimum growth (4 g/kg A. senticosus) revealed 820 differentially expressed genes and 44 significantly enriched pathways, especially lipid metabolism pathways such as unsaturated fatty acid and fatty acid metabolism. The transcript levels of five lipid metabolism-related genes were determined by quantitative real-time PCR. The results showed that 2–4 g/kg A. senticosus supplementation reduced the FADS2, ELOVL2, CYP24a, and PLPP3 transcript levels and 4 g/kg A. senticosus increased the DIO2 transcript level (P <0.05), leading to altered synthesis of TG and thyroxine and reduced fat deposition in the liver. Our results show that dietary A. senticosus affects the regulation of fat metabolism and promotes the growth of hybrid yellow catfish. A. senticosus is a healthy feed additive, and the appropriate dietary supplementation rate is 2–4 g/kg.
Collapse
Affiliation(s)
- Ming Xiao Li
- Wuxi Fisheries College, Nanjing Agricultural University, Jiangsu, Wuxi, China
| | - Jun Qiang
- Key Laboratory of Freshwater Fishes and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Jiangsu, Wuxi, China
| | - Jing Wen Bao
- Key Laboratory of Freshwater Fishes and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Jiangsu, Wuxi, China
| | - Yi Fan Tao
- Key Laboratory of Freshwater Fishes and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Jiangsu, Wuxi, China
| | - Hao Jun Zhu
- Key Laboratory of Freshwater Fishes and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Jiangsu, Wuxi, China
| | - Pao Xu
- Wuxi Fisheries College, Nanjing Agricultural University, Jiangsu, Wuxi, China
- Key Laboratory of Freshwater Fishes and Germplasm Resources Utilization, Ministry of Agriculture, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Jiangsu, Wuxi, China
- * E-mail:
| |
Collapse
|
34
|
Zhong F, Guan L, Lin H, Zhao M, Qin Y, Li Q, Yuan Z, Yang G, Gao L, Zhao J. Red Blood Cell Count: An Unrecognized Risk Factor for Nonalcoholic Fatty Liver Disease. Front Endocrinol (Lausanne) 2021; 12:760981. [PMID: 34950103 PMCID: PMC8688742 DOI: 10.3389/fendo.2021.760981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD) is becoming a global public health challenge. A convenient NAFLD indicator will greatly facilitate risk appraisal and prevention. As a readily available and inexpensive hematological index in routine clinical examinations, red blood cells (RBCs) are gaining increasing attention in many diseases, such as metabolic syndrome, but their association with NAFLD is unknown. METHODS This health management cohort study included 27,112 subjects (17,383 non-NAFLD and 9,729 NAFLD) with up to 5 years of follow-up (median 2.8 years). NAFLD was diagnosed by ultrasonography. NAFLD severity was categorized as mild, moderate, or severe. The generalized estimation equation (GEE), an extension of generalized linear models that allows for analysis of repeated measurements, was used to analyze the association between RBC count and NAFLD. RESULTS Overall, 4,332 of 17,383 (24.9%) subjects without NAFLD at baseline developed NAFLD. Incident NAFLD risk was positively associated with RBC count. After adjustment for hemoglobin and other confounders, the risk of incident NAFLD was 21%, 32%, and 51% higher in the second, third, and fourth RBC count quartiles, respectively, than in the lowest quartile. In 1,798 of 9,476 (19.0%) subjects with NAFLD at baseline, the severity of NAFLD increased. NAFLD progression risk increased progressively as RBC count increased (P for trend < 0.001). Every one-unit (1012 cells/L) increase in RBC count was associated with a 53% [OR 1.53 (95% CI 1.32-1.77)] increased risk for NAFLD progression. CONCLUSIONS Elevated RBC count was independently associated with a high risk of NAFLD incidence and progression. This finding revealed a convenient NAFLD risk indicator.
Collapse
Affiliation(s)
- Fang Zhong
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
- Shandong Institute of Endocrine and Metabolic Disease, Jinan, China
| | - Liying Guan
- Health Management Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Haiyan Lin
- Health Management Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Meng Zhao
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
- Shandong Institute of Endocrine and Metabolic Disease, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yiming Qin
- College of Chemical Engineering and Materials Science, Shandong Normal University, Jinan, China
| | - Qihang Li
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
- Shandong Institute of Endocrine and Metabolic Disease, Jinan, China
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Shandong University, Jinan, China
| | - Guang Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
- Shandong Institute of Endocrine and Metabolic Disease, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Clinical Medical Center of Endocrinology and Metabolism, Jinan, China
- Shandong Institute of Endocrine and Metabolic Disease, Jinan, China
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Jiajun Zhao,
| |
Collapse
|
35
|
Ishay Y, Rotnemer-Golinkin D, Ilan Y. The role of the sphingosine axis in immune regulation: A dichotomy in the anti-inflammatory effects between sphingosine kinase 1 and sphingosine kinase 2-dependent pathways. Int J Immunopathol Pharmacol 2021; 35:20587384211053274. [PMID: 34789044 PMCID: PMC8645305 DOI: 10.1177/20587384211053274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/21/2021] [Indexed: 01/05/2023] Open
Abstract
Background: Sphingosine kinase has been identified as playing a central role in the immune cascade, being a common mediator in the cellular response to a variety of signals. The different effects of sphingosine kinase 1 and 2 (SphK1 and SphK2, respectively) activity have not been completely characterized. Aim: To determine the different roles played by SphK1 and SphK2 in the regulation of immune-mediated disorders. Methods: Nine groups of mice were studied. Concanavalin A (ConA) injection was used to induce immune-mediated hepatitis. Mice were treated with SphK1 inhibitor (termed SphK-I) and SphK2 inhibitor (termed ABC294640), prior to ConA injection, and effects of treatment on liver enzymes, subsets of T lymphocytes, and serum levels of cytokines were observed. Results: While liver enzyme elevation was ameliorated by administration of SphK1 inhibitor, SphK2 inhibitor-treated mice did not show this tendency. A marked decrease in expression of CD25+ T-cells and Foxp+ T-cells was observed in mice treated with a high dose of SphK1 inhibitor. Alleviation of liver damage was associated with a statistically significant reduction of serum IFNγ levels in mice treated with SphK1 inhibitor and not in those treated with SphK2 inhibitor. Conclusions: Early administration of SphK1 inhibitor in a murine model of immune-mediated hepatitis alleviated liver damage and inflammation with a statistically significant reduction in IFN-γ levels. The data support a dichotomy in the anti-inflammatory effects of SphK1 and SphK2, and suggests that isoenzyme-directed therapies can improve the effect of targeting these pathways.
Collapse
Affiliation(s)
- Yuval Ishay
- Department of Medicine, Hadassah-Hebrew University Medical
Center, Jerusalem Israel
| | | | - Yaron Ilan
- Department of Medicine, Hadassah-Hebrew University Medical
Center, Jerusalem Israel
| |
Collapse
|
36
|
Wu X, Sakharkar MK, Wabitsch M, Yang J. Effects of Sphingosine-1-Phosphate on Cell Viability, Differentiation, and Gene Expression of Adipocytes. Int J Mol Sci 2020; 21:ijms21239284. [PMID: 33291440 PMCID: PMC7730007 DOI: 10.3390/ijms21239284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/27/2020] [Accepted: 12/04/2020] [Indexed: 11/16/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a highly potent sphingolipid metabolite, which controls numerous physiological and pathological process via its extracellular and intracellular functions. The breast is mainly composed of epithelial cells (mammary gland) and adipocytes (stroma). Adipocytes play an important role in regulating the normal functions of the breast. Compared to the vast amount studies on breast epithelial cells, the functions of S1P in breast adipocytes are much less known. Thus, in the current study, we used human preadipocyte cell lines SGBS and mouse preadipocyte cell line 3T3-L1 as in vitro models to evaluate the effects of S1P on cell viability, differentiation, and gene expression in adipocytes. Our results showed that S1P increased cell viability in SGBS and 3T3-L1 preadipocytes but moderately reduced cell viability in differentiated SGBS and 3T3-L1 adipocytes. S1P was also shown to inhibit adipogenic differentiation of SGBS and 3T3-L1 at concentration higher than 1000 nM. Transcriptome analyses showed that S1P was more influential on gene expression in differentiated adipocytes. Furthermore, our network analysis in mature adipocytes showed that the upregulated DEGs (differentially expressed genes) were related to regulation of lipolysis, PPAR (peroxisome proliferator-activated receptor) signaling, alcoholism, and toll-like receptor signaling, whereas the downregulated DEGs were overrepresented in cytokine-cytokine receptor interaction, focal adhesion, starch and sucrose metabolism, and nuclear receptors pathways. Together previous studies on the functions of S1P in breast epithelial cells, the current study implicated that S1P may play a critical role in modulating the bidirectional regulation of adipocyte-extracellular matrix-epithelial cell axis and maintaining the normal physiological functions of the breast.
Collapse
Affiliation(s)
- Xiyuan Wu
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada; (X.W.); (M.K.S.)
| | - Meena Kishore Sakharkar
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada; (X.W.); (M.K.S.)
| | - Martin Wabitsch
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Eythstr. 24, 89075 Ulm, Germany;
| | - Jian Yang
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada; (X.W.); (M.K.S.)
- Correspondence:
| |
Collapse
|
37
|
Li Q, Qian J, Li Y, Huang P, Liang H, Sun H, Liu C, Peng J, Lin X, Chen X, Peng H, Wang Z, Liu M, Shi Y, Yan H, Wei Y, Liao L, He Q, Huang X, Ruan F, Mao C, Zhou J, Wang K, Li C. Generation of sphingosine-1-phosphate by sphingosine kinase 1 protects nonalcoholic fatty liver from ischemia/reperfusion injury through alleviating reactive oxygen species production in hepatocytes. Free Radic Biol Med 2020; 159:136-149. [PMID: 32738398 DOI: 10.1016/j.freeradbiomed.2020.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/27/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Nonalcoholic fatty liver (NAFL) is emerging as a leading risk factor of hepatic ischemia/reperfusion (I/R) injury lacking of effective therapy. Lipid dyshomeostasis has been implicated in the hepatopathy of NAFL. Herein, we investigate the bioactive lipids that critically regulate I/R injury in NAFL. METHODS Lipidomics were performed to identify dysregulated lipids in mouse and human NAFL with I/R injury. The alteration of corresponding lipid-metabolizing genes was examined. The effects of the dysregulated lipid metabolism on I/R injury in NAFL were evaluated in mice and primary hepatocytes. RESULTS Sphingolipid metabolic pathways responsible for the generation of sphingosine-1-phosphate (S1P) were uncovered to be substantially activated by I/R in mouse NAFL. Sphingosine kinase 1 (Sphk1) was found to be essential for hepatic S1P generation in response to I/R in hepatocytes of NAFL mice. Sphk1 knockdown inhibited the hepatic S1P rise while accumulating ceramides in hepatocytes of NAFL mice, leading to aggressive hepatic I/R injury with upregulation of oxidative stress and increase of reactive oxygen species (ROS). In contrast, administration of exogenous S1P protected hepatocytes of NAFL mice from hepatic I/R injury. Clinical study revealed a significant activation of S1P generation by I/R in liver specimens of NAFL patients. In vitro studies on the L02 human hepatocytes consolidated that inhibiting the generation of S1P by knocking down SPHK1 exaggerated I/R-induced damage and oxidative stress in human hepatocytes of NAFL. CONCLUSIONS Generation of S1P by SPHK1 is important for protecting NAFL from I/R injury, which may serve as therapeutic targets for hepatic I/R injury in NAFL.
Collapse
Affiliation(s)
- Qingping Li
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianping Qian
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiyi Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Pengxiang Huang
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hanbiao Liang
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hang Sun
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Cuiting Liu
- Central Laboratory, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Peng
- Department of General Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinxin Lin
- The First Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Xuefang Chen
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongxian Peng
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zihuan Wang
- The First Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Meiqi Liu
- The First Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Yaru Shi
- The First Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongmei Yan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiran Wei
- The First Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Leyi Liao
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinghua He
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xixin Huang
- The First Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Fangyi Ruan
- The First Clinical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Cungui Mao
- Department of Medicine and Cancer Center, The State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Jie Zhou
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Kai Wang
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Chuanjiang Li
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
38
|
Mücke VT, Maria Schwarzkopf K, Thomas D, Mücke MM, Rüschenbaum S, Trebicka J, Pfeilschifter J, Zeuzem S, Lange CM, Grammatikos G. Serum Sphingosine-1-Phosphate Is Decreased in Patients With Acute-on-Chronic Liver Failure and Predicts Early Mortality. Hepatol Commun 2020; 4:1477-1486. [PMID: 33024917 PMCID: PMC7527696 DOI: 10.1002/hep4.1561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/17/2020] [Accepted: 06/10/2020] [Indexed: 12/30/2022] Open
Abstract
Sphingosine‐1‐phosphate (S1P) regulates pathophysiological processes, including liver regeneration, vascular tone control, and immune response. In patients with liver cirrhosis, acute deterioration of liver function is associated with high mortality rates. The present study investigated whether serum S1P concentrations are associated with disease severity in patients with chronic liver disease from compensated cirrhosis (CC), acute decompensation (AD), or acute‐on‐chronic liver failure (ACLF). From August 2013 to October 2017, patients who were admitted to the University Hospital Frankfurt with CC, AD, or ACLF were enrolled in our cirrhosis cohort study. Tandem mass spectrometry was performed on serum samples of 127 patients to assess S1P concentration. Our study comprised 19 patients with CC, 55 with AD, and 51 with ACLF, aged 29 to 76 years. We observed a significant decrease of S1P according to advanced liver injury from CC and AD up to ACLF (P < 0.001). S1P levels further decreased with progression to ACLF grade 3 (P < 0.05), and S1P highly inversely correlated with the Model for End‐Stage Liver Disease score (r = −0.508; P < 0.001). In multivariate analysis, S1P remained an independent predictor of 7‐day mortality with high diagnostic accuracy (area under the curve, 0.874; P < 0.001). Conclusion: In patients with chronic liver disease, serum S1P levels dramatically decreased with advanced stages of liver disease and were predictive of early mortality. Because S1P is a potent regulator of endothelial integrity and immune response, low S1P levels may significantly influence progressive multiorgan failure. Our data justify further elucidation of the diagnostic and therapeutic role of S1P in ACLF.
Collapse
Affiliation(s)
- Victoria T Mücke
- Departement of Internal Medicine 1 University Hospital Frankfurt Goethe University Frankfurt am Main Germany
| | - Katharina Maria Schwarzkopf
- Departement of Internal Medicine 1 University Hospital Frankfurt Goethe University Frankfurt am Main Germany
| | - Dominique Thomas
- Pharmazentrum Frankfurt Institute of Clinical Pharmacology Goethe University Frankfurt am Main Germany
| | - Marcus M Mücke
- Departement of Internal Medicine 1 University Hospital Frankfurt Goethe University Frankfurt am Main Germany
| | - Sabrina Rüschenbaum
- Department of Gastroenterology and Hepatology University Hospital Essen University of Duisburg-Essen Essen Germany
| | - Jonel Trebicka
- Departement of Internal Medicine 1 University Hospital Frankfurt Goethe University Frankfurt am Main Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt Institute of General Pharmacology and Toxicology Goethe University Frankfurt am Main Germany
| | - Stefan Zeuzem
- Departement of Internal Medicine 1 University Hospital Frankfurt Goethe University Frankfurt am Main Germany
| | - Christian M Lange
- Department of Gastroenterology and Hepatology University Hospital Essen University of Duisburg-Essen Essen Germany
| | - Georgios Grammatikos
- Departement of Internal Medicine 1 University Hospital Frankfurt Goethe University Frankfurt am Main Germany.,St. Luke's Hospital Thessaloniki Panorama Greece
| |
Collapse
|
39
|
Regulation of hepatic insulin signaling and glucose homeostasis by sphingosine kinase 2. Proc Natl Acad Sci U S A 2020; 117:24434-24442. [PMID: 32917816 PMCID: PMC7533871 DOI: 10.1073/pnas.2007856117] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatic insulin resistance is a chief pathogenic determinant in the development of type 2 diabetes, which is often associated with abnormal hepatic lipid regulation. Sphingolipids are a class of essential lipids in the liver, where sphingosine kinase 2 (SphK2) is a key enzyme in their catabolic pathway. However, roles of SphK2 and its related sphingolipids in hepatic insulin resistance remain elusive. Here we generate liver-specific Sphk2 knockout mice, demonstrating that SphK2 in the liver is essential for insulin sensitivity and glucose homeostasis. We also identify sphingosine as a bona fide endogenous inhibitor of hepatic insulin signaling. These findings provide physiological insights into SphK2 and sphingosine, which could be therapeutic targets for the management of insulin resistance and diabetes. Sphingolipid dysregulation is often associated with insulin resistance, while the enzymes controlling sphingolipid metabolism are emerging as therapeutic targets for improving insulin sensitivity. We report herein that sphingosine kinase 2 (SphK2), a key enzyme in sphingolipid catabolism, plays a critical role in the regulation of hepatic insulin signaling and glucose homeostasis both in vitro and in vivo. Hepatocyte-specific Sphk2 knockout mice exhibit pronounced insulin resistance and glucose intolerance. Likewise, SphK2-deficient hepatocytes are resistant to insulin-induced activation of the phosphoinositide 3-kinase (PI3K)-Akt-FoxO1 pathway and elevated hepatic glucose production. Mechanistically, SphK2 deficiency leads to the accumulation of sphingosine that, in turn, suppresses hepatic insulin signaling by inhibiting PI3K activation in hepatocytes. Either reexpressing functional SphK2 or pharmacologically inhibiting sphingosine production restores insulin sensitivity in SphK2-deficient hepatocytes. In conclusion, the current study provides both experimental findings and mechanistic data showing that SphK2 and sphingosine in the liver are critical regulators of insulin sensitivity and glucose homeostasis.
Collapse
|
40
|
Ji X, Yang L, Zhang Z, Zhang K, Chang N, Zhou X, Hou L, Yang L, Li L. Sphingosine 1‐phosphate/microRNA‐1249‐5p/MCP‐1 axis is involved in macrophage‐associated inflammation in fatty liver injury in mice. Eur J Immunol 2020; 50:1746-1756. [DOI: 10.1002/eji.201948351] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 05/02/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Xiaofang Ji
- Department of Cell Biology Municipal Laboratory for Liver Protection and Regulation of Regeneration Capital Medical University Beijing China
| | - Le Yang
- Department of Cell Biology Municipal Laboratory for Liver Protection and Regulation of Regeneration Capital Medical University Beijing China
| | - Zhi Zhang
- Department of Cell Biology Municipal Laboratory for Liver Protection and Regulation of Regeneration Capital Medical University Beijing China
| | - Kai Zhang
- Department of Cell Biology Municipal Laboratory for Liver Protection and Regulation of Regeneration Capital Medical University Beijing China
| | - Na Chang
- Department of Cell Biology Municipal Laboratory for Liver Protection and Regulation of Regeneration Capital Medical University Beijing China
| | - Xuan Zhou
- Department of Cell Biology Municipal Laboratory for Liver Protection and Regulation of Regeneration Capital Medical University Beijing China
| | - Lei Hou
- Department of Cell Biology Municipal Laboratory for Liver Protection and Regulation of Regeneration Capital Medical University Beijing China
| | - Lin Yang
- Department of Cell Biology Municipal Laboratory for Liver Protection and Regulation of Regeneration Capital Medical University Beijing China
| | - Liying Li
- Department of Cell Biology Municipal Laboratory for Liver Protection and Regulation of Regeneration Capital Medical University Beijing China
| |
Collapse
|
41
|
Blocking sphingosine 1-phosphate receptor 2 accelerates hepatocellular carcinoma progression in a mouse model of NASH. Biochem Biophys Res Commun 2020; 530:665-672. [PMID: 32768187 DOI: 10.1016/j.bbrc.2020.07.099] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 02/08/2023]
Abstract
The role of sphingosine 1-phosphate (S1P) and its sphingosine-1-phosphate receptors (S1PRs) in non-alcoholic steatohepatitis (NASH) is unclear. We aimed to analyze the role of S1P/S1PRs in a Melanocortin-4 receptor (Mc4r)-deficient NASH murine model using FTY720, the functional antagonist of S1PR1, S1PR3, S1PR4, and S1PR5, and JTE-013, the antagonist of S1PR2. We observed that, compared to that in the control, the mRNA of S1pr1 tended to decrease, whereas those of S1pr2 and S1pr3 significantly increased in Mc4r-knockout (KO) mice subjected to a Western diet (WD). While the fat area did not differ, fibrosis progression differed significantly between control mice and mice in which liver S1PRs were blocked. Lipidomic and metabolomic analysis of liver tissues showed that JTE-013-administered mice showed elevation of S-adenosyl-l-methionine level, which can induce aberrant methylation due to reduction in glycine N-methyltransferase (GNMT) and elevation in diacylglycerol (DG) and triacylglycerol (TG) levels, leading to increased susceptibility to hepatocellular carcinoma (HCC). These phenotypes are similar to those of Gnmt-KO mice, suggesting that blocking the S1P/S1PR2 axis triggers aberrant methylation, which may increase DG and TG, and hepatocarcinogenesis. Our observations that the S1P/S1PR2 axis averts HCC occurrence may assist in HCC prevention in NASH.
Collapse
|
42
|
Yu J, Dong J, Chen K, Ding Y, Yang Z, Lan T. Generation of mice with hepatocyte-specific conditional deletion of sphingosine kinase 1. Transgenic Res 2020; 29:419-428. [PMID: 32696422 DOI: 10.1007/s11248-020-00211-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
Abstract
SphK1 gene has different roles in various types of cells in liver diseases, but most studies are based on global knockout mice, which hampers the study on the cellular and molecular mechanisms of SphK1. In order to further study the role of SphK1 in liver, SphK1 conditional knockout mice were constructed. A liver-specific SphK1 gene knockout mouse model was constructed by the Cre/Loxp recombinant enzyme system. PCR technologies and western blotting were used to identified the elimination of SphK1 gene in hepatocytes. SphK1flox/flox mice were used as a control group to verify the effectiveness of SphK1 liver-specific knockout mice from the profile, pathology, and serology of mice. The ablation of SphK1 in hepatic parenchymal cells was demonstrated by fluorescent in situ hybridization and the contents of S1P and Sph were measured by ELISA kit. The genotypes of liver in SphK1 conditional knockout mice were different from that of other organs. The mRNA and protein levels of SphK1 in liver tissue of SphK1 conditional knockout mice were almost depleted by compared with SphK1flox/flox mice. Physiology and pathology showed no significant difference between SphK1 liver conditional knockout mice and SphK1flox/flox mice. Additionally, SphK1 was eliminated in hepatocytes, leading to the reduce of S1P content in hepatocytes and liver tissues and the increase of Sph content in hepatocytes. The model of SphK1 gene liver conditional knockout mice was successfully constructed, providing a tool for the study of the roles of SphK1 in hepatocyte and liver diseases.
Collapse
Affiliation(s)
- Jinfeng Yu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiale Dong
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Kangdi Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yaping Ding
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zhicheng Yang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Tian Lan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China. .,Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, 510006, China. .,Guangzhou Higher Education Mega Center, Guangdong Pharmaceutical University, 280 Wai Huan Dong Road, Guangzhou, 510006, China.
| |
Collapse
|
43
|
Yang T, Wang X, Yuan Z, Miao Y, Wu Z, Chai Y, Yu Q, Wang H, Sun L, Huang X, Zhang L, Jiang Z. Sphingosine 1-phosphate receptor-1 specific agonist SEW2871 ameliorates ANIT-induced dysregulation of bile acid homeostasis in mice plasma and liver. Toxicol Lett 2020; 331:242-253. [PMID: 32579994 DOI: 10.1016/j.toxlet.2020.06.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022]
Abstract
Dysregulated bile acid (BA) homeostasis is an extremely significant pathological phenomenon of intrahepatic cholestasis, and the accumulated BA could further trigger hepatocyte injury. Here, we showed that the expression of sphingosine-1-phosphate receptor 1 (S1PR1) was down-regulated by α-naphthylisothiocyanate (ANIT) in vivo and in vitro. The up-regulated S1PR1 induced by SEW2871 (a specific agonist of S1PR1) could improve ANIT-induced deficiency of hepatocyte tight junctions (TJs), cholestatic liver injury and the disrupted BA homeostasis in mice. BA metabolic profiles showed that SEW2871 not only reversed the disruption of plasma BA homeostasis, but also alleviated BA accumulation in the liver of ANIT-treated mice. Further quantitative analysis of 19 BAs showed that ANIT increased almost all BAs in mice plasma and liver, all of which were restored by SEW2871. Our data demonstrated that the top performing BAs were taurine conjugated bile acids (T-), especially taurocholic acid (TCA). Molecular mechanism studies indicated that BA transporters, synthetase, and BAs nuclear receptors (NRs) might be the important factors that maintained BA homeostasis by SEW2871 in ANIT-induced cholestasis. In conclusion, these results demonstrated that S1PR1 selective agonists might be the novel and potential effective agents for the treatment of intrahepatic cholestasis by recovering dysregulated BA homeostasis.
Collapse
Affiliation(s)
- Tingting Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Xue Wang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Zihang Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Yingying Miao
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Ziteng Wu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Yuanyuan Chai
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Qiongna Yu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Haiyan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Huang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
44
|
Xie Y, Chen L, Gao Y, Ma X, He W, Zhang Y, Zhang F, Fan Y, Gu L, Li P, Zhang X, Gou X. miR-363 suppresses the proliferation, migration and invasion of clear cell renal cell carcinoma by downregulating S1PR1. Cancer Cell Int 2020; 20:227. [PMID: 32536815 PMCID: PMC7288407 DOI: 10.1186/s12935-020-01313-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 06/01/2020] [Indexed: 12/23/2022] Open
Abstract
Background MicroRNAs (miRNAs) serve as important regulators of the tumorigenesis and progression of many human cancers. Therefore, we evaluated the biological function and underlying mechanism of miR-363 in clear cell renal cell carcinoma (ccRCC). Methods The expression of miR-363 in ccRCC tissues compared with adjacent normal renal tissues was detected by quantitative real-time polymerase chain reaction, and the association between miR-363 levels and prognosis of ccRCC patients was analyzed. The candidate target gene of miR-363 was determined by in silico analysis and luciferase reporter assays. The effects of miR-363 on the proliferation, migration and invasion of ccRCC cells in vitro were determined by MTS assay, colony formation assay, Transwell assay and wound healing assay. We also investigated the roles of miR-363 in vivo by a xenograft tumour model. The mechanism of miR-363 on the proliferation, migration and invasion of ccRCC was determined by gain- and loss-of-function analyses. Results we demonstrated that miR-363 expression was obviously downregulated in ccRCC tissues and that reduced miR-363 expression was correlated with poor disease-free survival (DFS) in ccRCC patients after surgery. S1PR1 expression was inversely correlated with the level of miR-363 in human ccRCC samples. Luciferase reporter assays suggested that S1PR1 was a direct functional target of miR-363. miR-363 downregulated S1PR1 expression and suppressed the proliferation, migration and invasion abilities of ccRCC cells in vitro and suppressed xenograft tumour growth in vivo. Importantly, miR-363 exerted its biological function by inhibiting S1PR1 expression in ccRCC cells, leading to the repression of ERK activation. Moreover, we found that the levels of downstream effectors of ERK, including PDGF-A, PDGF-B, and epithelial-mesenchymal transition (EMT)-related genes, were decreased after miR-363 overexpression. Conclusions Our results suggest that miR-363 acts as a tumour suppressor by directly targeting S1PR1 in ccRCC and may be a potential new therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Yongpeng Xie
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016 China
| | - Luyao Chen
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi China
| | - Yu Gao
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853 China
| | - Xin Ma
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853 China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016 China
| | - Yu Zhang
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853 China
| | - Fan Zhang
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853 China
| | - Yang Fan
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853 China
| | - Liangyou Gu
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853 China
| | - Pin Li
- Department of Pediatric Urology, Bayi Children's Hospital Affiliated to the Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xu Zhang
- Department of Urology, State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853 China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016 China
| |
Collapse
|
45
|
Park WJ, Song JH, Kim GT, Park TS. Ceramide and Sphingosine 1-Phosphate in Liver Diseases. Mol Cells 2020; 43:419-430. [PMID: 32392908 PMCID: PMC7264474 DOI: 10.14348/molcells.2020.0054] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/06/2020] [Accepted: 04/19/2020] [Indexed: 12/12/2022] Open
Abstract
The liver is an important organ in the regulation of glucose and lipid metabolism. It is responsible for systemic energy homeostasis. When energy need exceeds the storage capacity in the liver, fatty acids are shunted into nonoxidative sphingolipid biosynthesis, which increases the level of cellular ceramides. Accumulation of ceramides alters substrate utilization from glucose to lipids, activates triglyceride storage, and results in the development of both insulin resistance and hepatosteatosis, increasing the likelihood of major metabolic diseases. Another sphingolipid metabolite, sphingosine 1-phosphate (S1P) is a bioactive signaling molecule that acts via S1P-specific G protein coupled receptors. It regulates many cellular and physiological events. Since an increase in plasma S1P is associated with obesity, it seems reasonable that recent studies have provided evidence that S1P is linked to lipid pathophysiology, including hepatosteatosis and fibrosis. Herein, we review recent findings on ceramides and S1P in obesity-mediated liver diseases and the therapeutic potential of these sphingolipid metabolites.
Collapse
Affiliation(s)
- Woo-Jae Park
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 2999, Korea
| | - Jae-Hwi Song
- Department of Life Science, Gachon University, Seongnam 1310, Korea
| | - Goon-Tae Kim
- Department of Life Science, Gachon University, Seongnam 1310, Korea
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Seongnam 1310, Korea
| |
Collapse
|
46
|
Barron KA, Jeffries KA, Krupenko NI. Sphingolipids and the link between alcohol and cancer. Chem Biol Interact 2020; 322:109058. [PMID: 32171848 DOI: 10.1016/j.cbi.2020.109058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/20/2019] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Epidemiological evidence underscores alcohol consumption as a strong risk factor for multiple cancer types, with liver cancer being most commonly associated with alcohol intake. While mechanisms linking alcohol consumption to malignant tumor development are not fully understood, the likely players in ethanol-induced carcinogenesis are genotoxic stress caused by formation of acetaldehyde, increased oxidative stress, and altered nutrient metabolism, including the impairment of methyl transfer reactions. Alterations of sphingolipid metabolism and associated signaling pathways are another potential link between ethanol and cancer development. In particular, ceramides are involved in the regulation of cellular proliferation, differentiation, senescence, and apoptosis and are known to function as important regulators of malignant transformation as well as tumor progression. However, to date, the cross-talk between ceramides and alcohol in cancer disease is largely an open question and only limited data are available on this subject. Most studies linking ceramide to cancer considered liver steatosis as the underlying mechanism, which is not surprising taking into consideration that ceramide pathways are an integral part of the overall lipid metabolism. This review summarizes the latest studies pointing to ceramide as an important mediator of cancer-promoting effects of chronic alcohol consumption and underscores the necessity of understanding the role of sphingolipids and lipid signaling in response to alcohol in order to prevent and/or successfully manage diseases caused by alcohol.
Collapse
Affiliation(s)
| | | | - Natalia I Krupenko
- Department of Nutrition, UNC Chapel Hill, USA; Nutrition Research Institute, UNC Chapel Hill, USA.
| |
Collapse
|
47
|
Human antigen R: A potential therapeutic target for liver diseases. Pharmacol Res 2020; 155:104684. [PMID: 32045667 DOI: 10.1016/j.phrs.2020.104684] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 02/08/2023]
Abstract
Human antigen R (HuR), also known as HuA and embryonic lethal abnormal vision-like 1 (ELAVL1), is a ubiquitously expressed RNA binding protein and functions as an RNA regulator and mediates the expression of various proteins by diverse post-transcriptional mechanisms. HuR has been well characterized in the inflammatory responses and in the development of various cancers. The importance of HuR-mediated roles in cell signaling, inflammation, fibrogenesis and cancer development in the liver has attracted a great deal of attention. However, there is still a substantial gap between the current understanding of the potential roles of HuR in the progression of liver disease and whether HuR can be targeted for the treatment of liver diseases. In this review, we introduce the function and mechanistic characterization of HuR, and then focus on the physiopathological roles of HuR in the development of different liver diseases, including hepatic inflammation, alcoholic liver diseases, non-alcoholic fatty liver diseases, viral hepatitis, liver fibrosis and liver cancers. We also summarize existing approaches targeting HuR function. In conclusion, although characterizing the liver-specific HuR function and demonstrating the multi-level regulative networks of HuR in the liver are still required, emerging evidence supports the notion that HuR represents a potential therapeutic target for the treatment of chronic liver diseases.
Collapse
|
48
|
Fang H, Feng Q, Shi Y, Zhou J, Wang Q, Zhong L. Hepatic insulin resistance induced by mitochondrial oxidative stress can be ameliorated by sphingosine 1-phosphate. Mol Cell Endocrinol 2020; 501:110660. [PMID: 31759099 DOI: 10.1016/j.mce.2019.110660] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/22/2019] [Accepted: 11/19/2019] [Indexed: 12/21/2022]
Abstract
The bioactive lipid mediator sphingosine 1-phosphate (S1P) is considered to be involved in the development of insulin resistance (IR) via effects on oxidative stress; the mechanism however is not yet fully revealed. To this end, we investigated the role and mechanism of S1P on hepatic IR. We found that treatment of the normal human liver cell LO2 with 1000 nM insulin for 48 h reduced glucose uptake and increased serine phosphorylation of insulin receptor substrate-1, indicating a reduction in insulin receptor signaling. Moreover, the same concentration of insulin caused accumulation of reactive oxygen species (ROS) in the cytosol and mitochondria, and enhanced expression of the antioxidant transcription factor (Nrf2) and upregulated Nrf2 nuclear translocation. Using known inhibitors and donors of ROS (H2O2, ·O2-, ·OH), the results demonstrated the differential roles for the specific ROS in regulating IR in LO2 cells, with H2O2 having a more significant inhibitory role compared with ·O2- and ·OH. Cell treatment with S1P at 0.1-5.0 μM reversed the effects of high insulin concentrations on ROS generation, glucose uptake, and insulin signaling. H2O2 also reversed the beneficial effects of S1P in alleviating IR. These results show that H2O2 signaling plays a key determinant in hepatic IR induced by insulin. S1P can ameliorate hepatic IR by reducing mitochondrial ROS generation, and the possible anti-IR effect mechanism may be involved in H2O2 signaling.
Collapse
Affiliation(s)
- Hongjuan Fang
- Department of Endocrinology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Qiong Feng
- Chinese People's Liberation Army Center of Disease Control and Prevention, Beijing, 100071, China
| | - Yunxiang Shi
- Chinese People's Liberation Army Center of Disease Control and Prevention, Beijing, 100071, China
| | - Jiping Zhou
- Chinese People's Liberation Army Center of Disease Control and Prevention, Beijing, 100071, China
| | - Qiang Wang
- Chinese People's Liberation Army Center of Disease Control and Prevention, Beijing, 100071, China.
| | - Liyong Zhong
- Department of Endocrinology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
49
|
Hamdane N, Jühling F, Crouchet E, El Saghire H, Thumann C, Oudot MA, Bandiera S, Saviano A, Ponsolles C, Suarez AAR, Li S, Fujiwara N, Ono A, Davidson I, Bardeesy N, Schmidl C, Bock C, Schuster C, Lupberger J, Habersetzer F, Doffoël M, Piardi T, Sommacale D, Imamura M, Uchida T, Ohdan H, Aikata H, Chayama K, Boldanova T, Pessaux P, Fuchs BC, Hoshida Y, Zeisel MB, Duong FHT, Baumert TF. HCV-Induced Epigenetic Changes Associated With Liver Cancer Risk Persist After Sustained Virologic Response. Gastroenterology 2019; 156:2313-2329.e7. [PMID: 30836093 PMCID: PMC8756817 DOI: 10.1053/j.gastro.2019.02.038] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/14/2019] [Accepted: 02/27/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Chronic hepatitis C virus (HCV) infection is an important risk factor for hepatocellular carcinoma (HCC). Despite effective antiviral therapies, the risk for HCC is decreased but not eliminated after a sustained virologic response (SVR) to direct-acting antiviral (DAA) agents, and the risk is higher in patients with advanced fibrosis. We investigated HCV-induced epigenetic alterations that might affect risk for HCC after DAA treatment in patients and mice with humanized livers. METHODS We performed genome-wide ChIPmentation-based ChIP-Seq and RNA-seq analyses of liver tissues from 6 patients without HCV infection (controls), 18 patients with chronic HCV infection, 8 patients with chronic HCV infection cured by DAA treatment, 13 patients with chronic HCV infection cured by interferon therapy, 4 patients with chronic hepatitis B virus infection, and 7 patients with nonalcoholic steatohepatitis in Europe and Japan. HCV-induced epigenetic modifications were mapped by comparative analyses with modifications associated with other liver disease etiologies. uPA/SCID mice were engrafted with human hepatocytes to create mice with humanized livers and given injections of HCV-infected serum samples from patients; mice were given DAAs to eradicate the virus. Pathways associated with HCC risk were identified by integrative pathway analyses and validated in analyses of paired HCC tissues from 8 patients with an SVR to DAA treatment of HCV infection. RESULTS We found chronic HCV infection to induce specific genome-wide changes in H3K27ac, which correlated with changes in expression of mRNAs and proteins. These changes persisted after an SVR to DAAs or interferon-based therapies. Integrative pathway analyses of liver tissues from patients and mice with humanized livers demonstrated that HCV-induced epigenetic alterations were associated with liver cancer risk. Computational analyses associated increased expression of SPHK1 with HCC risk. We validated these findings in an independent cohort of patients with HCV-related cirrhosis (n = 216), a subset of which (n = 21) achieved viral clearance. CONCLUSIONS In an analysis of liver tissues from patients with and without an SVR to DAA therapy, we identified epigenetic and gene expression alterations associated with risk for HCC. These alterations might be targeted to prevent liver cancer in patients treated for HCV infection.
Collapse
Affiliation(s)
- Nourdine Hamdane
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Frank Jühling
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Emilie Crouchet
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Houssein El Saghire
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Christine Thumann
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Marine A. Oudot
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Simonetta Bandiera
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Antonio Saviano
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France,Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| | - Clara Ponsolles
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Armando Andres Roca Suarez
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Shen Li
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Naoto Fujiwara
- Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Atsushi Ono
- Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas,Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan,Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Irwin Davidson
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UDS, Illkirch, France
| | - Nabeel Bardeesy
- Center for Cancer Research, Massachusetts General Hospital; Departments of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Christian Schmidl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria,Regensburg Centre for Interventional Immunology (RCI) and University Medical Center of Regensburg, Regensburg, Germany
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria,Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria,Max Planck Institute for Informatics, Saarbrücken, Germany
| | - Catherine Schuster
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Joachim Lupberger
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - François Habersetzer
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| | - Michel Doffoël
- Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| | - Tullio Piardi
- General, Digestive, and Endocrine Surgery Unit, Hôpital Robert Debré, Centre Hospitalier Universitaire de Reims, Université de Reims Champagne-Ardenne, Reims, France
| | - Daniele Sommacale
- General, Digestive, and Endocrine Surgery Unit, Hôpital Robert Debré, Centre Hospitalier Universitaire de Reims, Université de Reims Champagne-Ardenne, Reims, France
| | - Michio Imamura
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takuro Uchida
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Aikata
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tujana Boldanova
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland,Division of Gastroenterology and Hepatology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Patrick Pessaux
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| | - Bryan C. Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Yujin Hoshida
- Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Mirjam B. Zeisel
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France,INSERM U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France
| | - François H. T. Duong
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Thomas F. Baumert
- INSERM U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France,Université de Strasbourg, Strasbourg, France,Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France,Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
50
|
Rohrbach TD, Asgharpour A, Maczis MA, Montefusco D, Cowart LA, Bedossa P, Sanyal AJ, Spiegel S. FTY720/fingolimod decreases hepatic steatosis and expression of fatty acid synthase in diet-induced nonalcoholic fatty liver disease in mice. J Lipid Res 2019; 60:1311-1322. [PMID: 31110049 DOI: 10.1194/jlr.m093799] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/16/2019] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), a leading cause of liver dysfunction, is a metabolic disease that begins with steatosis. Sphingolipid metabolites, particularly ceramide and sphingosine-1-phosphate (S1P), have recently received attention for their potential roles in insulin resistance and hepatic steatosis. FTY720/fingolimod, a prodrug for the treatment of multiple sclerosis, is phosphorylated in vivo to its active phosphorylated form by sphingosine kinase 2 and has been shown to interfere with the actions of S1P and to inhibit ceramide biosynthesis. Therefore, in this study we investigated the effects of FTY720 in a diet-induced animal model of NAFLD (DIAMOND) that recapitulates the hallmarks of the human disease. The oral administration of FTY720 to these mice fed a high-fat diet and sugar water improved glucose tolerance and reduced steatosis. In addition to decreasing liver triglycerides, FTY720 also reduced hepatic sphingolipid levels, including ceramides, monohexosylceramides, and sphingomyelins, particularly the C16:0 and C24:1 species, as well as S1P and dihydro-S1P. FTY720 administration decreased diet-induced fatty acid synthase (FASN) expression in DIAMOND mice without affecting other key enzymes in lipogenesis. FTY720 had no effect on the expression of SREBP-1c, which transcriptionally activates FASN. However, in agreement with the notion that the active phosphorylated form of FTY720 is an inhibitor of histone deacetylases, FTY720-P accumulated in the liver, and histone H3K9 acetylation was markedly increased in these mice. Hence, FTY720 might be useful for attenuating FASN expression and triglyceride accumulation associated with steatosis.
Collapse
Affiliation(s)
- Timothy D Rohrbach
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Amon Asgharpour
- Division of Gastroenterology, Hepatology, and Nutrition Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Melissa A Maczis
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, VA
| | - David Montefusco
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, VA
| | - L Ashley Cowart
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, VA.,Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, VA
| | - Pierre Bedossa
- Division of Gastroenterology, Hepatology, and Nutrition Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology, and Nutrition Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology Virginia Commonwealth University School of Medicine, Richmond, VA
| |
Collapse
|