1
|
Xia Q, Huang X, Li A, Zhuang Z, Zhou X, Yang Y, Zuo X, Liu Y, Sheng Y, Xu J, Cui Y. OASL activates MAPK to drive psoriatic pathogenesis: Astilbin targeting this axis improves metabolic-inflammation crosstalk. Life Sci 2025; 375:123698. [PMID: 40360089 DOI: 10.1016/j.lfs.2025.123698] [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: 02/19/2025] [Revised: 04/23/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
AIMS This study aimed to elucidate the role of oligoadenylate synthase-like protein (OASL) as a pivotal regulator integrating keratinocyte hyperproliferation, inflammation, and lipid metabolic dysregulation in psoriasis pathogenesis. MATERIALS AND METHODS Clinical analyses compared epidermal OASL expression levels between psoriasis patients and healthy individuals. Functional studies in HaCaT cells employed OASL knockdown and overexpression to assess effects on proliferation, inflammatory responses, and lipid metabolism. The JAK1-STAT1-OASL regulatory axis was investigated using the JAK1 inhibitor Upadacitinib. The natural flavonoid Astilbin was screened as an OASL inhibitor, and its therapeutic efficacy was evaluated in imiquimod-induced psoriatic mice. KEY FINDINGS OASL was significantly upregulated in psoriatic epidermis. Knockdown of OASL suppressed keratinocyte proliferation and inflammation, while its overexpression promoted hyperproliferation, inflammation, and lipid metabolic dysregulation via p38 MAPK pathway activation. Mechanistically, Upadacitinib reduced OASL expression by inhibiting STAT1, confirming the JAK1-STAT1-OASL axis. Astilbin markedly alleviated imiquimod-induced psoriasiform dermatitis in mice. SIGNIFICANCE This study reveals OASL's pivotal regulatory role in psoriasis and its associated pathways, providing novel therapeutic targets and a potential natural drug candidate. These findings establish a theoretical foundation for developing multi-targeted strategies against psoriasis.
Collapse
Affiliation(s)
- Qingyue Xia
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China.
| | - Xiaoyi Huang
- Institute of Dermatology and Department of Dermatology of the First Affiliated Hospital, Anhui Medical University, Hefei 230031, China
| | - Ang Li
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100029, China
| | - Zhou Zhuang
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Xinzhu Zhou
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; Peking University China-Japan Friendship School of Clinical Medicine Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Yue Yang
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Institute of Clinical Medical Sciences, Beijing 100029, China
| | - Xianbo Zuo
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yi Liu
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; Capital Medical University, Beijing 100069, China
| | - Yujun Sheng
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China; Institute of Dermatology and Department of Dermatology of the First Affiliated Hospital, Anhui Medical University, Hefei 230031, China.
| | - Jingkai Xu
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Yong Cui
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
2
|
Gruevska A, Leslie J, Perpiñán E, Maude H, Collins AL, Johnson S, Evangelista L, Sabey E, French J, White S, Moir J, Robinson SM, Alrawashdeh W, Thakkar R, Forlano R, Manousou P, Goldin R, Carling D, Hoare M, Thursz M, Mann DA, Cebola I, Posma JM, Safinia N, Oakley F, Hall Z. Spatial lipidomics reveals sphingolipid metabolism as anti-fibrotic target in the liver. Metabolism 2025; 168:156237. [PMID: 40127860 DOI: 10.1016/j.metabol.2025.156237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND AND AIMS Steatotic liver disease (SLD), which encompasses various causes of fat accumulation in the liver, is a major cause of liver fibrosis. Understanding the specific mechanisms of lipotoxicity, dysregulated lipid metabolism, and the role of different hepatic cell types involved in fibrogenesis is crucial for therapy development. METHODS We analysed liver tissue from SLD patients and 3 mouse models. We combined bulk/spatial lipidomics, transcriptomics, imaging mass cytometry (IMC) and analysis of published spatial and single-cell RNA sequencing (scRNA-seq) data to explore the metabolic microenvironment in fibrosis. Pharmacological inhibition of sphingolipid metabolism with myriocin, fumonisin B1, miglustat and D-PDMP was carried out in hepatic stellate cells (HSCs) and human precision cut liver slices (hPCLSs). RESULTS Bulk lipidomics revealed increased glycosphingolipids, ether lipids and saturated phosphatidylcholines in fibrotic samples. Spatial lipidomics detected >40 lipid species enriched within fibrotic regions, notably sphingomyelin (SM) 34:1. Using bulk transcriptomics (mouse) and analysis of published spatial transcriptomics data (human) we found that sphingolipid metabolism was also dysregulated in fibrosis at transcriptome level, with increased gene expression for ceramide and glycosphingolipid synthesis. Analysis of human scRNA-seq data showed that sphingolipid-related genes were widely expressed in non-parenchymal cells. By integrating spatial lipidomics with IMC of hepatic cell markers, we found excellent spatial correlation between sphingolipids, such as SM(34:1), and myofibroblasts. Inhibiting sphingolipid metabolism resulted in anti-fibrotic effects in HSCs and hPCLSs. CONCLUSIONS Our spatial multi-omics approach suggests cell type-specific mechanisms of fibrogenesis involving sphingolipid metabolism. Importantly, sphingolipid metabolic pathways are modifiable targets, which may have potential as an anti-fibrotic therapeutic strategy.
Collapse
Affiliation(s)
- Aleksandra Gruevska
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, University of Newcastle, Newcastle-upon-Tyne, United Kingdom
| | - Elena Perpiñán
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, United Kingdom
| | - Hannah Maude
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Amy L Collins
- Newcastle Fibrosis Research Group, Biosciences Institute, University of Newcastle, Newcastle-upon-Tyne, United Kingdom
| | - Sophia Johnson
- Newcastle Fibrosis Research Group, Biosciences Institute, University of Newcastle, Newcastle-upon-Tyne, United Kingdom
| | - Laila Evangelista
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Eleanor Sabey
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Jeremy French
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Steven White
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - John Moir
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Stuart M Robinson
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Wasfi Alrawashdeh
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Rohan Thakkar
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Roberta Forlano
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Pinelopi Manousou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Robert Goldin
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - David Carling
- MRC Laboratory of Medical Sciences, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Matthew Hoare
- Early Cancer Institute, University of Cambridge, Cambridge, United Kingdom
| | - Mark Thursz
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Derek A Mann
- Newcastle Fibrosis Research Group, Biosciences Institute, University of Newcastle, Newcastle-upon-Tyne, United Kingdom
| | - Inês Cebola
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Joram M Posma
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Niloufar Safinia
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, United Kingdom
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, University of Newcastle, Newcastle-upon-Tyne, United Kingdom; FibroFind, Unit 26/27, Baker's Yard, Christon Road, Newcastle upon Tyne, United Kingdom
| | - Zoe Hall
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| |
Collapse
|
3
|
Sasaki K, Ezaki H, Endo Y, Kudo D, Suenaga Y, Ayaori M, Sakurada M, Ikewaki K. Roles of HDL function and sphingosine-1-phosphate in vasospastic angina. Clin Chim Acta 2025; 574:120338. [PMID: 40320159 DOI: 10.1016/j.cca.2025.120338] [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: 02/17/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND High-density lipoprotein cholesterol (HDL-C) levels are often reduced in patients with vasospastic angina (VSA), but the relevance of HDL functionality to VSA pathogenesis remains unclear. Cholesterol uptake capacity (CUC), a novel cell-free assay reflecting HDL-mediated cholesterol efflux, offers a practical measure of HDL functionality. In parallel, sphingosine-1-phosphate (S1P), an HDL-associated bioactive sphingolipid with vasoprotective properties, may also contribute to VSA. This study aimed to evaluate CUC and vasodilatory HDL components, including S1P, in patients with VSA. METHODS AND RESULTS Seventy-seven patients, comprising 53 patients who underwent an acetylcholine (Ach) provocation test (32 VSA at diagnosis and 21 non-VSA) and an additional 24 VSA outpatients were included. Patients with VSA had higher triglyceride levels compared with non-VSA patients, but HDL-C levels were not different. Further analysis revealed that CUC was lower in VSA patients at diagnosis compared with non-VSA patients. Serum levels of sphingosine-1-phosphate (S1P), a sphingolipid associated with HDL, were elevated in the VSA group (1.74 ± 0.76 vs. 1.31 ± 0.49 µM; p < 0.01). In the VSA outpatient and Non-VSA groups, S1P levels in crude analysis were significantly associated with VSA (OR = 3.14, 95 % CI: 1.25-7.88, p = 0.01). This association remained significant across all adjusted models (Models 1-4). CONCLUSIONS The present study found that CUC was a novel indicator of vasospasm-related HDL dysfunctionality and that S1P is a promising biomarker for treated patients with VSA. The cholesterol efflux pathway and sphingolipid metabolism could contribute to the etiology of vasospasm. STUDY REGISTRATION This clinical study was registered with the University Hospital Medical Information Network Clinical Trials Registry (UMIN000020942).
Collapse
Affiliation(s)
- Kei Sasaki
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hirotaka Ezaki
- Department of Cardiology, Tokorozawa Heart Center, Tokorozawa, Saitama, Japan
| | - Yasuhiro Endo
- Division of Laboratory Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan.
| | - Daisuke Kudo
- Department of Cardiology, Tokorozawa Heart Center, Tokorozawa, Saitama, Japan
| | - Yumiko Suenaga
- Division of Anti-aging and Vascular Medicine, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Makoto Ayaori
- Department of Cardiology, Tokorozawa Heart Center, Tokorozawa, Saitama, Japan
| | - Masami Sakurada
- Department of Cardiology, Tokorozawa Heart Center, Tokorozawa, Saitama, Japan
| | - Katsunori Ikewaki
- Department of Cardiology, Tokorozawa Heart Center, Tokorozawa, Saitama, Japan
| |
Collapse
|
4
|
Zhenyu W, Mengyu L, Dongdong D, Jinyi H, Chuanmin Q, Hao Z, Xinjian L, Shenping Z, Wenshui X. A meta-analysis of genome-wide association studies revealed significant QTL and candidate genes for loin muscle area in three breeding pigs. Sci Rep 2025; 15:18758. [PMID: 40436882 PMCID: PMC12119988 DOI: 10.1038/s41598-025-00819-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/31/2025] [Indexed: 06/01/2025] Open
Abstract
Loin muscle area (LMA) is an important production trait in pigs and is highly correlated with lean meat percentage. However, the genetic architecture of LMA has not yet been fully elucidated. This study conducted genome-wide association studies (GWAS) and meta-analyses of LMA in Duroc (n = 337), Landrace (n = 662), and Yorkshire pigs (n = 3,176) using imputed whole-genome sequencing to identify new QTLs and candidate genes associated with LMA traits. A total of 108, 34, and 232 significant variants were identified in the Duroc, Landrace, and Yorkshire populations, respectively. The meta-analysis revealed 143 genome-wide significant SNPs and 276 suggestive SNPs, among which 213 were not identified in single population GWAS. Notably, 229 and 413 SNPs were located on SSC16 in the Yorkshire population and meta-analysis, respectively. Based on the 2-LOD drop-off interval, the SSC16 QTL in the Yorkshire population was further narrowed to a 679.835 kb interval (from 32.818 Mb to 33.498 Mb). The most significant variant within this QTL, 16_33228254 (P = 4.45 × 10-9), explained 1.11% phenotypic variance, representing a potential novel locus for LMA. Further bioinformatics analysis determined seven promising candidate genes (NDUFS4, ARL15, FST, ADAM12, DAB2, PLPP1, and SGMS2) with biological processes such as myoblast fusion and positive regulation of transforming growth factor beta receptor signaling pathway. Among them, ARL15 was previously reported in LMA studies, while the other six genes represent novel candidate genes. These findings reveal potential functional genes and pathways associated with LMA, providing valuable insights for future genetic improvement in pigs.
Collapse
Affiliation(s)
- Wang Zhenyu
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
| | - Li Mengyu
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
| | - Duan Dongdong
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
| | - Han Jinyi
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
| | - Qiao Chuanmin
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, 571100, Hainan, People's Republic of China
| | - Zhou Hao
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
| | - Li Xinjian
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, 571100, Hainan, People's Republic of China
| | - Zhou Shenping
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China.
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, 571100, Hainan, People's Republic of China.
| | - Xin Wenshui
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, Hainan, People's Republic of China.
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, 571100, Hainan, People's Republic of China.
| |
Collapse
|
5
|
Lin HM, Scheinberg T, Portman N, Kim RMN, Mellor R, Hunyh K, Faulkner AN, Mellett NA, Davis ID, Martin A, Sullivan D, Joshua A, McJannett M, Subhash V, Yip S, Azad AA, Marschner IC, North SA, McDermott RS, Chi KN, Stockler MR, Sweeney CJ, Meikle PJ, Horvath LG. Association of the circulating lipid panel, PCPro, with clinical outcomes in metastatic hormone-sensitive prostate cancer: post-hoc analysis of the ENZAMET Phase 3 randomised trial (ANZUP 1304). Ann Oncol 2025:S0923-7534(25)00732-X. [PMID: 40403846 DOI: 10.1016/j.annonc.2025.05.529] [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: 12/16/2024] [Revised: 04/08/2025] [Accepted: 05/07/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Enzalutamide significantly improves overall survival (OS) of patients with metastatic hormone- sensitive prostate cancer (mHSPC). However, ∼10% of patients will die within 2 years. PCPro is a plasma lipid panel associated with decreased OS in metastatic castration-resistant prostate cancer. In this study, we assessed the association between PCPro and clinical outcomes in mHSPC by performing a post-hoc analysis of ENZAMET, the landmark phase 3 trial comparing enzalutamide to non-steroidal anti-androgen (NSAA). PATIENTS AND METHODS PCPro status was determined by liquid chromatography-mass spectrometry analysis of plasma samples from 866 participants (77% of ENZAMET trial cohort), before treatment (n=866) and at first progression (n=282). Outcomes examined were OS and clinical progression-free survival (clinPFS). RESULTS Participants with a positive PCPro status at baseline (13.4%), had significantly shorter OS and clinPFS compared to those with a negative PCPro status (OS HR=1.81; clinPFS HR=1.65; p<0.0001). PCPro is an independent prognostic factor when modelled with key clinical prognostic factors (p<0.001). Enzalutamide (compared to NSAA) improved the OS of PCPro-negative participants (HR = 0.61, p<0.0001), but not the survival of PCPro-positive participants (HR=1.10, p=0.69; interaction p=0.024). Participants, who were PCPro-positive at progression, have shorter OS than those who were negative, irrespective of baseline status (median OS 24-28 months versus 42-45 months). CONCLUSION PCPro status is a prognostic biomarker and predictive of the lack of OS benefit from enzalutamide compared to NSAA in mHSPC. These findings provide a rationale for testing therapeutic agents that can modify circulating lipid profiles in mHSPC.
Collapse
Affiliation(s)
- H-M Lin
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Darlinghurst, NSW, Australia; Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia
| | - T Scheinberg
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; Chris O'Brien Lifehouse, Camperdown, NSW, Australia; University of Sydney, Camperdown, NSW, Australia
| | - N Portman
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Darlinghurst, NSW, Australia
| | - R M N Kim
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
| | - R Mellor
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Darlinghurst, NSW, Australia; Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | - K Hunyh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Cardiovascular Research Translation and Implementation, La Trobe University, Bundoora, VIC, Australia
| | - A N Faulkner
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - N A Mellett
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - I D Davis
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; Eastern Health Clinical School, Monash University, VIC, Australia; Cancer Services, Eastern Health, Melbourne, Australia
| | - A Martin
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; Centre for Clinical Research, University of Queensland, Herston, QLD, Australia
| | - D Sullivan
- New South Wales Health Pathology, Department of Chemical Pathology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - A Joshua
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Darlinghurst, NSW, Australia; Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; Kinghorn Cancer Centre, St Vincent's Hospital, Darlinghurst, NSW, Australia
| | - M McJannett
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia
| | - V Subhash
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia
| | - S Yip
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - A A Azad
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - I C Marschner
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - S A North
- University of Alberta, Edmonton, AB, Canada
| | - R S McDermott
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; St Vincent's University Hospital Dublin, Ireland; Cancer Trials Ireland, Dublin, Ireland
| | - K N Chi
- British Columbia Cancer Agency, Vancouver, BC, Canada
| | - M R Stockler
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; Chris O'Brien Lifehouse, Camperdown, NSW, Australia; University of Sydney, Camperdown, NSW, Australia; National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Camperdown, NSW, Australia
| | - C J Sweeney
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; South Australian Immunogenomics Cancer Institute, Adelaide, SA, Australia
| | - P J Meikle
- University of Sydney, Camperdown, NSW, Australia; Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - L G Horvath
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; School of Clinical Medicine, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Darlinghurst, NSW, Australia; Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Australia; Chris O'Brien Lifehouse, Camperdown, NSW, Australia; University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
6
|
Naja K, Anwardeen N, Albagha O, Elrayess MA. Lipid Subclasses Differentiate Insulin Resistance by Triglyceride-Glucose Index. Metabolites 2025; 15:342. [PMID: 40422918 DOI: 10.3390/metabo15050342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2025] [Revised: 05/10/2025] [Accepted: 05/20/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Insulin resistance is a key driver of metabolic syndrome and related disorders, yet its underlying metabolic alterations remain incompletely understood. The Triglyceride-Glucose (TyG) index is an emerging, accessible marker for insulin resistance, with growing evidence supporting its clinical utility. This study aimed to characterize the metabolic profiles associated with insulin resistance using the TyG index in a large, population-based cohort, and to identify metabolic pathways potentially implicated in insulin resistance. METHODS Here, we conducted a cross-sectional study using data from the Qatar Biobank, including 1255 participants without diabetes classified as insulin-sensitive or insulin-resistant based on TyG index tertiles. Untargeted serum metabolomics profiling was performed using high-resolution mass spectrometry. Our statistical analyses included orthogonal partial least squares discriminate analysis and linear models. RESULTS Distinct metabolic signatures differentiated insulin-resistant from insulin-sensitive participants. Phosphatidylethanolamines, phosphatidylinositols, and phosphatidylcholines, were strongly associated with insulin resistance, while plasmalogens and sphingomyelins were consistently linked to insulin sensitivity. CONCLUSIONS Lipid-centric pathways emerge as potential biomarkers and therapeutic targets for the early detection and personalized management of insulin resistance and related metabolic disorders. Longitudinal studies are warranted to validate causal relationships.
Collapse
Affiliation(s)
- Khaled Naja
- Biomedical Research Center, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Najeha Anwardeen
- Biomedical Research Center, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Omar Albagha
- Division of Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Doha P.O. Box 34110, Qatar
- Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| | - Mohamed A Elrayess
- Biomedical Research Center, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
- College of Medicine, QU Health, Qatar University, Doha P.O Box 2713, Qatar
| |
Collapse
|
7
|
Chen W, Kullmann S, Rhea EM. Expanding the understanding of insulin resistance in brain and periphery. Trends Endocrinol Metab 2025:S1043-2760(25)00099-2. [PMID: 40393910 DOI: 10.1016/j.tem.2025.04.010] [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: 11/22/2024] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/22/2025]
Abstract
Insulin resistance is a central feature of metabolic disorders such as type 2 diabetes (T2D). While studies on this disorder have largely been linked to glucose metabolism and intracellular signaling, recent advances reveal that insulin resistance extends beyond traditional glucose regulatory pathways, impacting multiple organs including the brain, contributing to cognitive dysfunction and neurodegenerative diseases such as Alzheimer's disease (AD). This opinion revisits insulin resistance through molecular, cellular, and systemic perspectives, emphasizing the intersection between peripheral and brain insulin resistance (BIR), the role of the blood-brain barrier (BBB), and emerging biomarkers. Furthermore, we integrate insights from multi-omics and neuroimaging studies to refine our understanding, advocating for a broader perspective that informs early detection and intervention in metabolic and neurodegenerative diseases.
Collapse
Affiliation(s)
- Wenqiang Chen
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev, Denmark.
| | - Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; Department of Internal Medicine, Division of Diabetology, Endocrinology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany; German Center for Diabetes Research, München-Neuherberg, Germany
| | - Elizabeth M Rhea
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
8
|
Jiang J, Gao Y, Wang J, Huang Y, Yang R, Zhang Y, Ma Y, Wen Y, Luo G, Zhang S, Cao Y, Yu M, Wang Q, Hu S, Wang K, Guo X, Gonzalez FJ, Liu Y, Liu H, Xie Q, Xie C. Hepatic sphingomyelin phosphodiesterase 3 promotes steatohepatitis by disrupting membrane sphingolipid metabolism. Cell Metab 2025; 37:1119-1136.e13. [PMID: 40015281 DOI: 10.1016/j.cmet.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/16/2024] [Accepted: 01/17/2025] [Indexed: 03/01/2025]
Abstract
Metabolic-dysfunction-associated steatohepatitis (MASH) remains a major health challenge. Herein, we identify sphingomyelin phosphodiesterase 3 (SMPD3) as a key driver of hepatic ceramide accumulation through increasing sphingomyelin hydrolysis at the cell membrane. Hepatocyte-specific Smpd3 gene disruption or pharmacological inhibition of SMPD3 alleviates MASH, whereas reintroducing SMPD3 reverses the resolution of MASH. Although healthy livers express low-level SMPD3, lipotoxicity-induced DNA damage suppresses sirtuin 1 (SIRT1), triggering an upregulation of SMPD3 during MASH. This disrupts membrane sphingomyelin-ceramide balance and promotes disease progression by enhancing caveolae-dependent lipid uptake and extracellular vesicle secretion from steatotic hepatocytes to exacerbate inflammation and fibrosis. Consequently, SMPD3 acts as a central hub integrating key MASH hallmarks. Notably, we discovered a bifunctional agent that simultaneously activates SIRT1 and inhibits SMPD3, which shows significant therapeutic potential in MASH treatment. These findings suggest that inhibition of hepatic SMPD3 restores membrane sphingolipid metabolism and holds great promise for developing novel MASH therapies.
Collapse
Affiliation(s)
- Jie Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yuqing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiang Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Lingang Laboratory, Shanghai 200444, China
| | - Yan Huang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Rong Yang
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yongxin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuandi Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingquan Wen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Gongkai Luo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shurui Zhang
- Lingang Laboratory, Shanghai 200444, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yutang Cao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Minjun Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qinxue Wang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Shulei Hu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kanglong Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Frank J Gonzalez
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yameng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China.
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China.
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210046, China.
| |
Collapse
|
9
|
Dai D, Gao L, Pan Y, Chen C, Ma K, Zhang H, Wu S, Qi G, Wang J. Eggshell depigmentation in the late phase of production is associated with altered Microbiota and Metabolism of the uterus in laying hens. Poult Sci 2025; 104:105258. [PMID: 40367565 DOI: 10.1016/j.psj.2025.105258] [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: 02/06/2025] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
The significant depigmentation of brown eggshells occurs in the in the late-phase laying hens, which directly affects consumer acceptance. However, the biological mechanism of eggshell depigmentation based on uterine metabolism has not been elucidated. In this study, a total of 4 group were as follows: 1) 65-week-old laying hens with normal color; 2) 65-week-old laying hens with light color; 3) 80-week-old laying hens with normal color; 4) 80-week-old laying hens with light color. Variations in the pigment contents, uterine antioxidant capacity, uterine microbiota, and uterine metabolomics were examined in current study. Results showed that significantly decreased L* values and increased a* and b* values were observed in the depigmentation group (P < 0.05). The protoporphyrin IX content of the uterus with eggshell depigmentation was significantly decreased in 65-week-old laying hens (P < 0.05). Uterine MDA content was significantly increased in the depigmentation groups at 65 and 80 weeks of age, accompanied by reduced SOD and increased IgA levels (P > 0.05). The abundance of Proteobacteria and Campilobacterota was markedly reduced in the uterus with eggshell depigmentation, whereas Firmicutes was elevated at 65 weeks of age (P < 0.05). Further, Psychrobacte as biomarkers can accurately distinguish between normal color and depigmentation in eggshells (AUC = 0.91). A total of 51 differential metabolites were significantly enriched in the down-regulated sphingolipid metabolism, linoleic acid metabolism, citrate cycle, oxidative phosphorylation, PPAR signaling pathway, FoxO signaling pathway, and apoptosis at 65 weeks of age (P < 0.05). Meanwhile, there were 82 differential metabolites were significantly up-regulated at 80 weeks of age, which mainly enriched in up-regulated linoleic acid metabolism, purine metabolism, and pentose phosphate pathway (P < 0.05). These findings elucidate the specific metabolic mechanisms responsible for eggshell depigmentation in 65- and 80-week-old laying hens, contributing to the improvement of eggshell depigmentation by the precise nutritional modulation in the late-phase laying hens.
Collapse
Affiliation(s)
- Dong Dai
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China
| | - Libing Gao
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China
| | - Yingli Pan
- Beijing Huadu Yukou Poultry Industry Co., Ltd, Beijing 101200, PR China
| | - Chaojiang Chen
- Beijing Huadu Yukou Poultry Industry Co., Ltd, Beijing 101200, PR China
| | - Kaixuan Ma
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China
| | - Haijun Zhang
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China
| | - Shugeng Wu
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China
| | - Guanghai Qi
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China
| | - Jing Wang
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China.
| |
Collapse
|
10
|
Yan M, Hu J, Wang L, Zhang T, Xiao Z, Yuan Y, Yue T. Metabolic profiling of abdominal subcutaneous adipose tissue reveals effects of apple polyphenols for reversing high-fat diet induced obesity in C57BL/6 J mice. Food Chem 2025; 473:143055. [PMID: 39879748 DOI: 10.1016/j.foodchem.2025.143055] [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: 10/02/2024] [Revised: 01/07/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
Apple polyphenols (APP) can reduce obesity. However, the effects of APP on abdominal subcutaneous adipose tissue (aSAT) at metabolic level were unclear. In this study, 5-week APP intervenes were conducted on 10-week high-fat diet (HFD) feeding mice with doses of 200 and 500 mg/kg b.w./day, followed by ultra-high-performance liquid chromatography-mass spectrometry based untargeted metabolomics analysis. As expected, APP obviously reversed aSAT weight and index, as well as activities of myeloperoxidase, glutathione peroxidase, superoxide dismutase and catalase. Euclidean distance between HFD and normal chow diet (NCD) group was shortened. 64 and 127 differential metabolites were found in 200 and 500 mg/kg b.w./day group, with 12 and 13 changed pathways, respectively. Specifically, APP restored glycolysis, tricarboxylic acid cycle, amino acid metabolism, and lipid metabolism as dose-dependent manner. Finally, glucose-6-phosphate, xanthine and tyrosine were selected as critical junctures. Collectively, these findings underscore the potential of APP in reversing molecular alterations in aSAT.
Collapse
Affiliation(s)
- Min Yan
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Jinpeng Hu
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Lan Wang
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Ting Zhang
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Zhengcao Xiao
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Yahong Yuan
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China.
| | - Tianli Yue
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China.
| |
Collapse
|
11
|
Gao S, Liu B, Yuan S, Quan Y, Song S, Jin W, Wang Y, Wang Y. Cross-talk between signal transduction systems and metabolic networks in antibiotic resistance and tolerance. Int J Antimicrob Agents 2025; 65:107479. [PMID: 40024604 DOI: 10.1016/j.ijantimicag.2025.107479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/19/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
The comprehensive antibiotic resistance of pathogens signifies the oneset of the "post-antibiotic era", and the myriad treatment challenges posed by "superbugs" have emerged as the primary threat to human health. Recent studies indicate that bacterial resistance and tolerance development are mediated at the metabolic level by various signalling networks (e.g., quorum sensing systems, second messenger systems, and two-component systems), resulting in metabolic rearrangements and alterations in bacterial community behaviour. This review focuses on current research, highlighting the intrinsic link between signalling and metabolic networks in bacterial resistance and tolerance.
Collapse
Affiliation(s)
- Shuji Gao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Baobao Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Shuo Yuan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Yingying Quan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Shenao Song
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Wenjie Jin
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China.
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China; Henan Provincial Engineering Research Center for Detection and Prevention and Control of Emerging Infectious Diseases in Livestock and Poultry, Luoyang, China.
| |
Collapse
|
12
|
Yamada C, Akkaoui J, Morozov A, Movila A. Role of Canonical and Non-Canonical Sphingolipids and their Metabolic Enzymes in Bone Health. Curr Osteoporos Rep 2025; 23:21. [PMID: 40266422 PMCID: PMC12018623 DOI: 10.1007/s11914-025-00908-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE OF REVIEW This review summarizes the recently published scientific evidence regarding the role of enzymes engaged in de novo anabolic biosynthesis, catabolic, and salvage pathways of ceramide bioactive sphingolipids in bone dynamics and skeletal health. RECENT FINDINGS Ceramides are precursors for bioactive sphingolipids, including sphingosine, sphingosine-1-phosphate, and others. Studies of bone metabolism and bone-related cells demonstrated that ceramide and sphingosine-1-phosphate control levels of bone remodeling and resorption generated by osteoblasts and osteoclasts. Multiple published studies demonstrated the critical role of enzymes in regulating the ceramide/sphingosine-1-phosphate ratio relative to bone physiology and the promotion of inflammatory osteolysis. Accordingly, emerging evidence suggests that targeting sphingolipid metabolism has the potential to alleviate inflammatory osteolysis and accelerate bone regeneration. Therefore, this study aimed to discuss current knowledge about crosstalk between sphingolipids and their metabolic enzymes within osteoclast and osteoblast coupling in bone remodeling and pathogenic osteolysis. This review highlights the complexity of de novo sphingolipid biosynthesis and knowledge gaps in bone physiology and pathology. We also discuss the importance of canonical and non-canonical mammalian and bacterial-derived sphingolipids relative to bone health.
Collapse
Affiliation(s)
- Chiaki Yamada
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN, USA
| | - Juliet Akkaoui
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Alexandr Morozov
- Institute of Zoology, Moldova State University, Chisinau, Republic of Moldova
- Medpark International Hospital, Chisinau, Republic of Moldova
| | - Alexandru Movila
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA.
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
13
|
Zhao M, Bian R, Xu X, Zhang J, Zhang L, Zheng Y. Sphingolipid Metabolism and Signalling Pathways in Heart Failure: From Molecular Mechanism to Therapeutic Potential. J Inflamm Res 2025; 18:5477-5498. [PMID: 40291458 PMCID: PMC12034266 DOI: 10.2147/jir.s515757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
Sphingolipids are essential components of cell membranes and lipoproteins. They are synthesized de novo in the endoplasmic reticulum and subsequently undergo various enzymatic modifications in different organelles, giving rise to a diverse range of biologically active compounds. These molecules play a critical role in regulating cell growth, senescence, migration, apoptosis, and signaling. In recent years, the sphingolipid metabolic pathway has been recognized as a key factor in heart failure (HF) pathophysiology. Abnormal levels of sphingolipid metabolites, such as ceramide (Cer) and sphingomyelin (SM), contribute to oxidative stress and inflammatory responses, ultimately promoting cardiomyocyte apoptosis. Conversely, sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P) regulate vascular function and influence cardiac remodeling. Additionally, enzymes such as diacylglycerol acyltransferase 1 (DGAT1) and sphingosine-1-phosphate lyase 1 (SGPL1) modulate cardiac lipid metabolism. Given their role in HF progression, monitoring sphingolipid alterations offers potential as valuable biomarkers for assessing disease severity, prognosis, and diagnosis. Given the complexity of sphingolipid metabolism and its involvement in diverse regulatory biological processes, a comprehensive understanding of its roles at both the cellular and organismal levels in physiopathology remains incomplete. Therefore, this review aims to explore the physiological functions, regulatory mechanisms, and therapeutic potential of sphingolipid metabolism. It will summarize the specific molecular mechanisms driving key pathological processes in HF, including ventricular remodeling, myocardial fibrosis, vascular dysfunction, and metabolic disorders. Finally, the review will highlight targeted sphingolipid metabolites as potential therapeutic strategies, offering new insights into HF diagnosis and treatment, with the goal of advancing adjunctive clinical therapies.
Collapse
Affiliation(s)
- Meng Zhao
- The First Clinical Medical College of Henan University of Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
- Joint Formula and Syndrome Research Laboratory of Guangzhou University of Chinese Medicine & Zhengzhou Hospital of Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| | - Rutao Bian
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| | - Xuegong Xu
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| | - Junpeng Zhang
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| | - Li Zhang
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| | - Yi Zheng
- Department of Cardiology, Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou, Henan Province, People’s Republic of China
| |
Collapse
|
14
|
Ai X, Zhang Q, Ma Q, Fang M, Zhu K, Cai Y, Yang Q, Zhang L. Transcriptomics and metabolomics analysis of the pathogenesis of a novel hyperlipidemia-susceptible rat strain. Exp Anim 2025; 74:160-172. [PMID: 39496388 PMCID: PMC12044359 DOI: 10.1538/expanim.24-0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
Wistar-SD Hypercholesterolemia (WSHc) Rat is a novel hyperlipidemia-susceptible rat that we discovered and bred earlier, which can be used as an ideal animal model for the study of non-alcoholic fatty liver disease (NAFLD). However, its pathogenesis of hyperlipidemia and genetic and biological characteristics need to be further investigated. In the current study, WSHc rats were fed a high-fat diet (HFD) and standard chow (SC), with age-matched Wistar rats as the control group undergoing the same treatment, followed by serum lipid level measurement. It was found that HFD-fed WSHc rats developed dyslipidemia. Transcriptomic analysis was performed to detect genes associated with cholesterol metabolism in the liver, and 119 differentially expressed genes were discovered through bioinformatics analysis and molecular biology verification. Additionally, Srebf1 was identified as a HUB gene and Nr1d1 as an independent key gene using the protein-protein interaction network and one-cluster clustering analysis. The two genes had also been further validated in molecular biology experiments and were consistent with transcriptomic results. Serum lipid metabolomics analysis identified 7 lipid subclasses and 84 lipid molecules using UHPLC-Q-TOF/MS. There were 62 and 70 lipid molecules with significant differences in the metabolic profiles of serum lipid mediators in the WSHc+HFD group compared to the WSHc+SC and Wistar+HFD groups, respectively, and the differential metabolites were mainly produced via sphingolipid and glycerophospholipid metabolism. In sum, the hypercholesterolemia model can be established with WSHc rats after the HFD induction, and the pathogenesis involves the Srebf1 and Nr1d1 genes and the sphingolipid and glycerophospholipid metabolism pathways.
Collapse
Affiliation(s)
- Xiufeng Ai
- Animal Experimental Research Center/Institute of Comparative Medicine, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, P.R. China
| | - Qian Zhang
- Animal Experimental Research Center/Institute of Comparative Medicine, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, P.R. China
| | - Quanxin Ma
- Key laboratory of silkworm and bee resource utilization and innovation of Zhejiang Province, College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou, Zhejiang 310058, P.R. China
- Hangzhou Lifutai Biotechnology Co., LTD, 9 Juyuan Road, Binjiang District, Hangzhou, Zhejiang 310051, P.R. China
| | - Mingsun Fang
- Animal Experimental Research Center/Institute of Comparative Medicine, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, P.R. China
| | - Keyan Zhu
- Animal Experimental Research Center/Institute of Comparative Medicine, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, P.R. China
| | - Yueqin Cai
- Animal Experimental Research Center/Institute of Comparative Medicine, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, P.R. China
| | - Qinqin Yang
- Department of Experimental Animals, Zhejiang Academy of Traditional Chinese Medicine, 132 Tianmushan Road, Xihu District, Hangzhou, Zhejiang 310007, P.R. China
| | - Lizong Zhang
- Animal Experimental Research Center/Institute of Comparative Medicine, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
15
|
Hannun YA, Merrill AH, Luberto C. The Bioactive Sphingolipid Playbook. A Primer for the Uninitiated as well as Sphingolipidologists. J Lipid Res 2025:100813. [PMID: 40254066 DOI: 10.1016/j.jlr.2025.100813] [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: 02/04/2025] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025] Open
Abstract
Sphingolipids and glycosphingolipids are among the most structurally diverse and complex compounds in the mammalian metabolome. They are well known to play important roles in biological architecture, cell-cell communication and cellular regulation, and for many biological processes, multiple sphingolipids are involved. Thus, it is not surprising that untargeted genetic/transcriptomic/pharmacologic/metabolomic screens have uncovered changes in sphingolipids and sphingolipid genes/proteins while studying physiological and pathological processes. Consequently, with increasing frequency, both targeted and untargeted mass spectrometry methodologies are being used to conduct sphingolipidomic analyses. Interpretation of such large data sets and design of follow-up experiments can be daunting for investigators with limited expertise with sphingolipids (and sometimes even for someone well-versed in sphingolipidology). Therefore, this review gives an overview of essential elements of sphingolipid structure and analysis, metabolism, functions, and roles in disease, and discusses some of the items to consider when interpreting lipidomics data and designing follow-up investigations.
Collapse
Affiliation(s)
- Yusuf A Hannun
- Departments of Biochemistry, Medicine, and the Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| | - Alfred H Merrill
- School of Biological Sciences and the Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Chiara Luberto
- Department of Physiology and Biophysics, and the Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
16
|
Liu C, Zheng X, Ji J, Zhu X, Liu X, Liu H, Guo L, Ye K, Zhang S, Xu YJ, Sun X, Zhou W, Wong HLX, Tian Y, Qian H. The carotenoid torularhodin alleviates NAFLD by promoting Akkermanisa muniniphila-mediated adenosylcobalamin metabolism. Nat Commun 2025; 16:3338. [PMID: 40199868 PMCID: PMC11978934 DOI: 10.1038/s41467-025-58500-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
Torularhodin, a unique carotenoid, confers beneficial effects on nonalcoholic fatty liver disease (NAFLD). However, the precise mechanism underlying its therapeutic effects remains unknown. Here, we report that torularhodin alleviates NAFLD in male mice by modulating the gut microbiota. Additionally, transplanting fecal microbiota from torularhodin-treated mice to germ-free mice also improves NAFLD. Mechanistically, torularhodin specifically enriches the abundance of Akkermansia muciniphila, which alleviates NAFLD by promoting the synthesis of adenosylcobalamin. Utilizing a human gastrointestinal system and a colonic organoid model, we further demonstrate that adenosylcobalamin confers protective effects against NAFLD through reducing ceramides, a well-known liver damaging compound, and this effect is mediated by inhibition of the hypoxia-inducible factor 2α pathway. Notably, we construct electrospun microsphere-encapsulated torularhodin, which facilitates the slow release of torularhodin in the colon. Together, our findings indicate the therapeutic potential of microbial utilization of carotenoids, such as torularhodin, for treating NAFLD.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaojiao Zheng
- Center for Translational Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Ji
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xuan Zhu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, China
| | - Xiaoning Liu
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore, 138673, Singapore
| | - He Liu
- Department of Cardiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lichun Guo
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Kun Ye
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Shuang Zhang
- Analysis and Testing Center, Jiangnan University, Wuxi, China
| | - Yong-Jiang Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiulan Sun
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Weibiao Zhou
- Department of Food Science and Technology, National University of Singapore, Singapore, 117542, Singapore
- National University of Singapore (Suzhou) Research Institute, Suzhou, Jiangsu, China
| | | | - Yaoqi Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
- Analysis and Testing Center, Jiangnan University, Wuxi, China.
| | - He Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
17
|
Liu T, Zhang M, Xie Q, Gu J, Zeng S, Huang D. Unveiling the Antiobesity Mechanism of Sweet Potato Extract by Microbiome, Transcriptome, and Metabolome Analyses in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7807-7821. [PMID: 39989409 DOI: 10.1021/acs.jafc.4c13173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
This study aimed to elucidate the antiobesity mechanisms of sweet potato extract (SPE) through biochemical, gut microbiome, liver transcriptome, and metabolome analyses. Administration of SPE to high-fat-diet-fed mice significantly reduced body weight gain, serum low-density lipoprotein cholesterol, hepatic lipid accumulation, and adipocyte hypertrophy, which were closely linked to gut microbiome composition. SPE notably increased the abundance of Eubacterium_coprostanoligenes_group_unclassified and decreased that of Kineothrix, both of which were strongly associated with short-chain fatty acid (SCFA) production. LC-QTOF-MS analysis identified resin glycoside compounds from SPE with reduced levels in mouse feces, suggesting their utilization in vivo. SPE also promoted dietary fat excretion. Liver transcriptomic and metabolomic profiling revealed that SPE may exert antiobesity effects by modulating the bile-sphingolipid metabolism, which was closely correlated with the reshaped gut microbiomes and SCFAs. These findings provide new insights into the antiobesity effects and mechanisms of SPE.
Collapse
Affiliation(s)
- Tiange Liu
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, Jiangsu, China
| | - Min Zhang
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, Jiangsu, China
| | - Qingtong Xie
- Department of Food Science and Technology, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore
| | - Jia Gu
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, Jiangsu, China
| | - Shunjiang Zeng
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, Jiangsu, China
| | - Dejian Huang
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou 215123, Jiangsu, China
- Department of Food Science and Technology, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore
| |
Collapse
|
18
|
Li Y, Zhong S, Huang S, Zhong W, Zheng B, Guo Q, Liu J, Guo X, Su R. Application of metabolomics in the classification of traditional Chinese medicine syndromes in rheumatoid arthritis. Clin Rheumatol 2025; 44:1493-1504. [PMID: 40011356 DOI: 10.1007/s10067-025-07373-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/01/2025] [Accepted: 02/13/2025] [Indexed: 02/28/2025]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is frequently treated with traditional Chinese medicine (TCM), where patients are classified into distinct syndromes, such as heat-dampness syndrome (HD) and kidney-liver deficiency syndrome (KLD). However, an objective and systematic approach to differentiate these TCM syndromes remains lacking. This study is aimed at analyzing serum metabolomics to identify differential metabolites and pathways associated with HD and GS syndromes in RA patients and at evaluating their potential as diagnostic biomarkers. METHODS Serum samples from RA patients classified into HD and KLD groups were analyzed using metabolomics. Partial least squares discriminant analysis was employed to identify significant metabolites, while pathway analysis was conducted using the Kyoto Encyclopedia of Genes and Genomes database. Receiver operating characteristic (ROC) curve analysis was performed to assess the diagnostic potential of key metabolites. RESULTS Fifteen differential metabolites and two perturbed pathways-sphingolipid and D-amino acid metabolism-were identified between the KLD and HD groups. Notably, several metabolites, including C17-sphinganine and leucyl-alanine, demonstrated high diagnostic efficacy, with area under the curve (AUC) values exceeding 0.90. Correlation analysis revealed significant associations between certain metabolites and clinical indices, further substantiating their role in syndrome differentiation. CONCLUSION This study presents a comprehensive analysis of serum metabolites in RA patients under different TCM syndromes. The identified metabolites hold potential as biomarkers for distinguishing HD and KLD groups, paving the way for more objective and evidence-based diagnostic approaches in TCM. Key Points • Differential metabolites were identified in the serum of RA patients with heat-dampness syndrome and kidney-liver deficiency syndrome, with their metabolic pathways primarily involving sphingolipid metabolism and D-amino acid metabolism. • Serum metabolites demonstrate high efficacy in distinguishing RA patients with different TCM syndromes. • Significant correlations were observed between serum differential metabolites and clinical indicators in RA patients with varying TCM syndromes.
Collapse
Affiliation(s)
- Yao Li
- Department of Laboratory Medicine, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Engineering and Technology Research Center for Innovative and Precise Inspection Technology, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Shuqi Zhong
- Department of Laboratory Medicine, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Shengchun Huang
- Department of Laboratory Medicine, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Wanying Zhong
- Department of Laboratory Medicine, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Baolin Zheng
- Nephrology and Rheumatology Department, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Qihong Guo
- Nephrology and Rheumatology Department, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Jihong Liu
- Prevention and Treatment Center, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Xueyan Guo
- Department of Laboratory Medicine, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
- Foshan Engineering and Technology Research Center for Innovative and Precise Inspection Technology, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Rong Su
- Department of Laboratory Medicine, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China.
- Foshan Engineering and Technology Research Center for Innovative and Precise Inspection Technology, Foshan Hospital of Traditional Chinese Medicine, The Eighth Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan, Guangdong, China.
| |
Collapse
|
19
|
Wu R, Yu H, Xu J, Tan Z, Lan Y, Shi D. Effects of acute low intensity aerobics and blueberry juice on arterial stiffness in young adults. NPJ Sci Food 2025; 9:47. [PMID: 40169604 PMCID: PMC11962078 DOI: 10.1038/s41538-025-00408-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
Arterial stiffness, a cardiovascular disease (CVD) predictor starting from youth, is under-researched in young adults. Low-intensity aerobic exercise (LAE) is generally more accessible than higher-intensity exercise and may be more sustainable for young individuals. Blueberries, renowned for vascular health benefits, may reduce arterial stiffness. This study examines the effects of LAE and blueberry juice on arterial stiffness in 48 young adults. Participants were randomized into LAE, low-, mid-, or high-volume blueberry juice (LB, MB, HB), LAE + LB, LAE + MB, LAE + HB, and control groups. Arterial stiffness was measured at baseline and at 15-, 30-, 45-, and 60 min post-intervention. Blood samples were collected pre-intervention and 30-min post-intervention for metabolomic analysis. Repeated ANOVA revealed LAE + MB significantly reduced arterial stiffness. Metabolomic analysis revealed changes in linoleic acid, sphingolipid, phenylalanine, nicotinate and nicotinamide, glycerophospholipid, and lysine degradation metabolic pathways. These findings suggest a feasible exercise-diet strategy for CVD prevention in young adults and provide metabolic insights into the mechanisms.
Collapse
Affiliation(s)
- Ruisi Wu
- Changchun Normal University, Changchun, Jilin, 130032, China
| | - Huali Yu
- Key Laboratory of Molecular Epigenetics, Ministry of Education and Institute of Cytology and Genetics, Northeast Normal University, Changchun, 130024, China
| | - Jiayuan Xu
- Tonghua Changbaishan Wild Economic Plant Research Institute, Tonghua, Jilin, 134100, China
| | - Zhiqiang Tan
- Tonghua Changbaishan Wild Economic Plant Research Institute, Tonghua, Jilin, 134100, China
| | - Yongsheng Lan
- Changchun Normal University, Changchun, Jilin, 130032, China.
| | - Dongfang Shi
- Changchun Normal University, Changchun, Jilin, 130032, China.
| |
Collapse
|
20
|
Park K, Shin KO, Kim YI, Nielsen-Scott AL, Mainzer C, Celli A, Bae Y, Chae S, An H, Choi Y, Park JH, Park SH, Hwang JT, Kang SG, Wakefield JS, Arron ST, Holleran WM, Mauro TM, Elias PM, Uchida Y. Sphingosine-1-Phosphate-Cathelicidin Axis Plays a Pivotal Role in the Development of Cutaneous Squamous Cell Carcinoma. J Invest Dermatol 2025; 145:854-863.e6. [PMID: 39218144 DOI: 10.1016/j.jid.2024.08.008] [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: 10/12/2023] [Revised: 07/10/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is a common skin cancer caused by mutagenesis resulting from excess UVR or other types of oxidative stress. These stressors also upregulate the production of a cutaneous innate immune element, cathelicidin antimicrobial peptide (CAMP), through endoplasmic reticulum stress-initiated, sphingosine-1-phosphate (S1P) signaling pathway. Although CAMP has beneficial antimicrobial activities, it also can be proinflammatory and procarcinogenic. We addressed whether and how S1P-induced CAMP production leads to cSCC development. Our study demonstrated that (i) CAMP expression is increased in cSCC cells and skin from patients with cSCC; (ii) S1P levels are elevated in cSCC cells, whereas inhibition of S1P production attenuates CAMP-stimulated cSCC growth; (iii) exogenous CAMP stimulates cSCC but not normal human keratinocyte growth; (iv) blockade of FPRL1 protein, a CAMP receptor, attenuates cSCC growth as well as the growth and invasion of cSCC cells mediated by CAMP into an extracellular matrix-containing fibroblast substrate; (v) FOXP3+ regulatory T-cell (which decreases antitumor immunity) levels increase in cSCC skin; and (vi) CAMP induces endoplasmic reticulum stress in cSCC cells. Together, the endoplasmic reticulum stress-S1P-CAMP axis forms a vicious circle, creating a favorable environment for cSCC development, that is, cSCC growth and invasion impede anticancer immunity.
Collapse
Affiliation(s)
- Kyungho Park
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea; Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA.
| | - Kyong-Oh Shin
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea; LaSS, Chuncheon, Republic of Korea
| | - Young-Il Kim
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Anna L Nielsen-Scott
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Carine Mainzer
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Anna Celli
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Yoojin Bae
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea
| | - Seungwoo Chae
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea
| | - Hahyun An
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea
| | - Yerim Choi
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea; LaSS, Chuncheon, Republic of Korea
| | - Jae-Ho Park
- Personalized Diet Research Group, Korea Food Research Institute, Jeonju, Republic of Korea
| | - Soo-Hyun Park
- Personalized Diet Research Group, Korea Food Research Institute, Jeonju, Republic of Korea
| | - Jin-Taek Hwang
- Personalized Diet Research Group, Korea Food Research Institute, Jeonju, Republic of Korea; Department of Food Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Seung Goo Kang
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Joan S Wakefield
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Sarah T Arron
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Walter M Holleran
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Theodora M Mauro
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Peter M Elias
- Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - Yoshikazu Uchida
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Republic of Korea; Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Republic of Korea; Department of Dermatology, School of Medicine, University of California, San Francisco, San Francisco, California, USA; San Francisco VA Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA.
| |
Collapse
|
21
|
Villella VR, Castaldo A, Scialò F, Castaldo G. How Effectively Can Oxidative Stress and Inflammation Be Reversed When CFTR Function Is Pharmacologically Improved? Antioxidants (Basel) 2025; 14:310. [PMID: 40227282 PMCID: PMC11939277 DOI: 10.3390/antiox14030310] [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: 01/27/2025] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
A critical challenge in the age of advanced modulator therapies is to understand and determine how effectively chronic oxidative stress and oxidative stress-induced inflammation can be reversed and physiological balance restored when CFTR function is pharmacologically improved. The triple therapy with elexacaftor-tezacaftor-ivacaftor (ETI) suggests that CFTR activity in individuals with at least one F508del mutation can be partially restored to about 50% of normal levels. Although incomplete, the partial recovery of CFTR function has been shown to drastically lower sputum pathogen content, enhance microbiome diversity, and lower inflammation markers within the first year of treatment in adolescents and adults with cystic fibrosis. However, despite these advancements, residual airway infection, oxidative stress and inflammation persist, with levels similar to other chronic lung conditions, like non-CF bronchiectasis. This persistence highlights the necessity for innovative antioxidant and anti-inflammatory treatments, in particular for individuals with advanced lung disease. To address this issue, emerging multi-omics technologies offer valuable tools to investigate the impact of modulator therapies on various molecular pathways. By analyzing changes in gene expression, epigenetic modifications, protein profiles and metabolic processes in airway-derived samples, it could be possible to uncover the mechanisms driving persistent oxidative stress and inflammation. These insights could pave the way for identifying new therapeutic targets to fully restore airway health and overall physiological balance.
Collapse
Affiliation(s)
| | - Alice Castaldo
- SC di Pneumologia e UTSIR, AORN Santobono-Pausilipon, 80122 Naples, Italy;
- Dipartimento di Scienze Mediche Traslazionali, Sezione di Pediatria, Università di Napoli Federico II, 80131 Naples, Italy
| | - Filippo Scialò
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy; (V.R.V.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| | - Giuseppe Castaldo
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy; (V.R.V.); (G.C.)
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| |
Collapse
|
22
|
Gaggini M, Suman AF, Vassalle C. Ceramide in Coronary Artery Disease: Troublesome or Helpful Future Tools in the Assessment of Risk Prediction and Therapy Effectiveness? Metabolites 2025; 15:168. [PMID: 40137133 PMCID: PMC11943838 DOI: 10.3390/metabo15030168] [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: 02/04/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
Lipids are a complex entity of different molecules, among which ceramides (Cers), ubiquitous sphingolipids with remarkable biological activity, can represent a potential additive biomarker that can be used to better understand the underlying mechanisms which drive the onset and development of atherosclerotic damage and plaque vulnerability and facilitate coronary disease management, as possible risk/prognostic biomarkers and targets for therapeutic intervention. Accordingly, this review aims to discuss the available results on the role Cersplay in contributing to atherosclerosis development and acute coronary event precipitation, their impact on complications and adverse prognosis, as well as the impact of treatment options in modulating Cerlevels.
Collapse
Affiliation(s)
- Melania Gaggini
- Institute of Clinical Physiology, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy; (M.G.); (A.F.S.)
| | - Adrian Florentin Suman
- Institute of Clinical Physiology, National Research Council, Via G. Moruzzi 1, 56124 Pisa, Italy; (M.G.); (A.F.S.)
| | - Cristina Vassalle
- Fondazione CNR-Regione Toscana G Monasterio, Via G. Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
23
|
Zhao F, Shao M, Li M, Li T, Zheng Y, Sun W, Ni C, Li L. Sphingolipid metabolites involved in the pathogenesis of atherosclerosis: perspectives on sphingolipids in atherosclerosis. Cell Mol Biol Lett 2025; 30:18. [PMID: 39920588 PMCID: PMC11804087 DOI: 10.1186/s11658-024-00679-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/17/2024] [Indexed: 02/09/2025] Open
Abstract
Atherosclerosis, with its complex pathogenesis, is a leading underlying cause of many cardiovascular diseases, which are increasingly prevalent in the population. Sphingolipids play an important role in the development of atherosclerosis. Key metabolites and enzymes in sphingolipid metabolism influence the pathogenesis of atherosclerosis in a variety of ways, including inflammatory responses and oxidative stress. Thus, an investigation of sphingolipid metabolism-related metabolites and key enzymes may provide novel insights and treatment targets for atherosclerosis. This review discusses various mechanisms and research progress on the relationship between various sphingolipid metabolites, related enzymes, and atherosclerosis. Finally, we look into the future research direction of phytosphingolipids.
Collapse
Affiliation(s)
- Fufangyu Zhao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Mingyan Shao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mingrui Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tianxing Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yanfei Zheng
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wenlong Sun
- Institute of Biomedical Research, School of Life Sciences, Shandong University of Technology, Zibo, 255000, Shandong, China.
| | - Cheng Ni
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Lingru Li
- National Institute of Traditional Chinese Medicine Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
24
|
Zhang YH, Xie R, Dai CS, Gao HW, Zhou G, Qi TT, Wang WY, Wang H, Cui YM. Thyroid hormone receptor-beta agonist HSK31679 alleviates MASLD by modulating gut microbial sphingolipids. J Hepatol 2025; 82:189-202. [PMID: 39181210 DOI: 10.1016/j.jhep.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND & AIMS As the first approved medication for metabolic dysfunction-associated steatohepatitis (MASH), the thyroid hormone receptor-β (THR-β) agonist MGL-3196 (resmetirom) has garnered much attention as a liver-directed, bioactive oral drug. However, studies on MGL-3196 have also identified remarkable heterogeneity of individual clinical efficacy and its interference with gut microbiota in host hepatoenteral circulation remains to be elucidated. METHODS We compared MASH attenuation by MGL-3196 and its derivative drug HSK31679 between germ-free (GF) and specific-pathogen free (SPF) mice to evaluate the role of gut microbiota. Then cross-omics analyses of microbial metagenome, metabolome and single-cell RNA-sequencing were applied to a randomized, double-blind, placebo-controlled multiple ascending dose cohort receiving HSK31679 treatment (n = 32) or placebo (n = 8), to comprehensively investigate the altered gut microbiota metabolism and circulating immune signatures. RESULTS HSK31679 outperformed MGL-3196 in ameliorating MASH diet-induced steatohepatitis of SPF mice but not GF mice. In the multiple ascending dose cohort of HSK31679, the relative abundance of B. thetaiotaomicron was significantly enriched, impairing glucosylceramide synthase (GCS)-catalyzed monoglucosylation of microbial Cer(d18:1/16:0) and Cer(d18:1/24:1). In contrast to the non-inferior effect of MGL-3196 and HSK31679 on MASH resolution in GFBTΔGCS mice, HSK31679 led to superior benefit on steatohepatitis in GFBTWT mice, due to its steric hindrance of R123 and Y401 of gut microbial GCS. For participants with high fecal GCS activity, the administration of 160 mg HSK31679 induced a shift in peripheral compartments towards an immunosuppressive niche, characterized by decreased CD8α+ dendritic cells and MINCLE+ macrophages. CONCLUSIONS This study provided novel insights into the gut microbiota that are key to the efficacy of HSK31679 treatment, revealing microbial GCS as a potential predictive biomarker in MASH, as well as a new target for further microbiota-based treatment strategies for MASH. IMPACT AND IMPLICATIONS Remarkable heterogeneity in individual clinical efficacy of thyroid hormone receptor-β agonists and their interferences with the microbiome in host hepatoenteral circulation are poorly understood. In our current germ-free mouse models and a randomized, double-blind, multiple-dose cohort study, we identified microbial glucosylceramide synthase as a key mechanistic node in the resolution of metabolic dysfunction-associated steatohepatitis. Microbial glucosylceramide synthase activity could be a predictive biomarker of response to HSK31679 treatment or a new target for microbiota-based therapeutics in metabolic dysfunction-associated steatohepatitis.
Collapse
Affiliation(s)
- Yu-Hang Zhang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, 100191, China; Department of Pharmacy, Peking University First Hospital, Beijing, 100034, China
| | - Ran Xie
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, 100191, China; Department of Pharmacy, Peking University First Hospital, Beijing, 100034, China
| | - Chen-Shu Dai
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, 100191, China; Department of Pharmacy, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hong-Wei Gao
- Biomarker Technologies Corporation, Beijing, 101300, China
| | - Gan Zhou
- Department of Pharmacy, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Tian-Tian Qi
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, 100191, China
| | - Wen-Yu Wang
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, 230032, China.
| | - Yi-Min Cui
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, 100191, China; Department of Pharmacy, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
25
|
McKenna S, Jung KI, Wolf JJ, Seo YJ, Hahm B. Multiple sphingolipid-metabolizing enzymes modulate influenza virus replication. Virology 2025; 603:110367. [PMID: 39754863 PMCID: PMC11793951 DOI: 10.1016/j.virol.2024.110367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 01/06/2025]
Abstract
The sphingolipid network is sustained principally by the balance of bioactive sphingolipid molecules and their regulation by sphingolipid-metabolizing enzymes. The components in the lipid system display key functions in numerous cellular and disease conditions including virus infections. During the COVID-19 pandemic, there was a fruitful effort to use an inhibitor that blocks the activity of sphingosine kinase (SphK) 2 to cure the devastating disease. Support for the inhibitor came from pre-clinical research on influenza where the inhibitor demonstrated effective protection of mice from influenza-induced morbidity and mortality. This highlights the importance of basic and translational research on the sphingolipid system for improving human health. Multiple sphingolipid-metabolizing enzymes have been reported to regulate influenza virus replication and propagation. In this review, the emphasis is placed on the roles of these enzymes that impact influenza virus life cycle and the conceivable mechanisms for the interplay between influenza virus and the sphingolipid pathway.
Collapse
Affiliation(s)
- Savannah McKenna
- Departments of Surgery & Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA
| | - Kwang Il Jung
- Departments of Surgery & Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA
| | - Jennifer J Wolf
- Departments of Surgery & Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA
| | - Young-Jin Seo
- Department of Life Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| | - Bumsuk Hahm
- Departments of Surgery & Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
26
|
Zhao JQ, Zhou QQ, Liu K, Li P, Jiang Y, Li HJ. Lipidomics reveals the lipid-lowering and hepatoprotective effects of Celosia Semen on high-fat diet-induced NAFLD mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118922. [PMID: 39389395 DOI: 10.1016/j.jep.2024.118922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Celosia Semen (CS) serves as a traditional Chinese medicine (TCM) for promoting liver health and enhancing vision, with extensive clinical applications. Triterpenoid saponins represent the primary active components of CS, with the highest concentration of Celosin I (CI) detected. The urgent need for effective NAFLD treatments motivated us assess the beneficial effects of total saponins from CS (TSCS) and CI. AIMS OF THE STUDY To investigate the therapeutic effects of TSCS and CI on NAFLD and its underlying molecular mechanisms. MATERIALS AND METHODS The impact of TSCS and CI on NAFLD was evaluated through in vitro and in vivo methodologies, utilizing high-fat diet (HFD) and palmitic acid/oleic acid modeling on C57BL/6J mice and AML12 cells, respectively. Biochemical analysis, H&E and Oil red O staining were used to characterize the lipid-lowering and hepatoprotective activities of TSCS and CI. Lipidomics discerned the impact of TSCS and CI interventions on liver lipid composition, distribution and alteration in NFALD mice. RT-qPCR and western blotting detected the influence of TSCS and CI on genes linked to de novo lipogenesis, fat calculation uptake, oxidation and esterification. The docking analysis anticipated the interaction of six major triterpenoid saponins within TSCS with SREBP1. RESULTS TSCS and CI markedly diminished lipid accumulation induced by high fat both in vivo and in vitro. TSCS and CI also mitigated hepatic steatosis and liver injury induced by HFD through the reduction of TC, TG, FAs, ALT, and AST, even at minimal dose of 25 mg/kg. Lipidomics indicated that TSCS and CI had the potential to modulate the lipid metabolism network, rectify lipid metabolic dysregulation induced by NAFLD, decrease the levels of harmful lipids, and elevate the levels of advantageous lipids. Furthermore, TSCS and CI exhibited a strong affinity to SREBP1, thereby might directly influence the expression of SREBP1 and a cascade of essential enzymes involved in de novo lipogenesis, and finally resulting in a diminished synthesis of novel lipids. CONCLUSION TSCS and CI were confirmed firstly as key active components of CS in amending hepatic steatosis and mitigate liver damage in NAFLD, outlining the preliminary mechanism. They warrant further exploration as drug candidates for NAFLD treatment, especially in light of the current shortage of medications and limited therapeutic options.
Collapse
Affiliation(s)
- Jin-Quan Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, China
| | - Qi-Qi Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, China
| | - Ke Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, China
| | - Yan Jiang
- Nanjing Forestry University, Nanjing, 210037, China.
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Avenue, Nanjing, 211198, China.
| |
Collapse
|
27
|
Wang Y, Xue Y, Xu H, Zhu Q, Qin K, He Z, Huang A, Mu M, Tao X. Pediococcus acidilactici Y01 reduces HFD-induced obesity via altering gut microbiota and metabolomic profiles and modulating adipose tissue macrophage M1/M2 polarization. Food Funct 2025; 16:554-569. [PMID: 39699275 DOI: 10.1039/d4fo04301d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Obesity-related metabolic syndrome is intimately associated with infiltrated adipose tissue macrophages (ATMs), gut microbiota, and metabolic disorders. Pediococcus acidilactici holds the potential to mitigate obesity; however, there exist strain-specific functionalities and diverse mechanisms, which deserve extensive exploration. This study aims to explore the potential of P. acidilactici Y01, isolated from traditional sour whey, in alleviating HFD-induced metabolic syndrome in mice and elucidating its underlying mechanism. The results showed that P. acidilactici Y01 could inhibit the increase of body weight gain, the deposition of fat, lipid disorders and chronic low-grade inflammation, improve glucose tolerance and insulin resistance, and could reduce adipose tissue inflammation by decreasing M1-type ATMs and increasing M2-type ATMs. Meanwhile, P. acidilactici Y01 significantly increased the abundance of potentially beneficial intestinal bacteria, such as Akkermansia, Alistipes, Bifidobacterium, Lachnospiraceae_NK4A136_group, Lactobacillus, norank_f__Muribaculaceae, and Parabacteroides, and partially restored the levels of metabolites, such as phosphatidylcholines, glycerophosphocholines, sphingolipids and unsaturated fatty acids. The fecal microbiota transplantation experiment demonstrated that P. acidilactici Y01 ameliorated obesity-related metabolic syndrome by modulating the polarization of M1/M2 ATMs mediated by gut microbiota. Overall, as a dietary supplement, P. acidilactici Y01 has good potential in the prevention and treatment of obesity.
Collapse
Affiliation(s)
- Yujing Wang
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and, Technology, Huainan 232000, China.
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
- Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, Hefei, China
| | - Yu Xue
- School of Medicine, Department of Medical Frontier Experimental Center, Anhui University of Science and Technology, Huainan 232001, China
| | - Huan Xu
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and, Technology, Huainan 232000, China.
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
- Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, Hefei, China
| | - Qian Zhu
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
| | - Kaili Qin
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and, Technology, Huainan 232000, China.
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
- Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, Hefei, China
| | - Zhonglei He
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
| | - Aixiang Huang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Min Mu
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and, Technology, Huainan 232000, China.
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
- Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, Hefei, China
| | - Xinrong Tao
- Joint Research Center for Occupational Medicine and Health of IHM, Anhui University of Science and, Technology, Huainan 232000, China.
- School of Public Health, Anhui University of Science and Technology, Hefei 231131, China
- Key Laboratory of Industrial Dust Prevention and Control, Occupational Safety and Health, Ministry of Education, Anhui University of Science and Technology, Huainan 232001, China
- Anhui Institute of Occupational Safety and Health, Anhui University of Science and Technology, Hefei, China
| |
Collapse
|
28
|
Lin L, Huang Y, Qian S, Chen L, Sun H. Genetically predicted causal link between the plasma lipidome and pancreatic diseases: a bidirectional Mendelian randomization study. Front Nutr 2025; 11:1466509. [PMID: 39882037 PMCID: PMC11774697 DOI: 10.3389/fnut.2024.1466509] [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: 07/18/2024] [Accepted: 12/16/2024] [Indexed: 01/31/2025] Open
Abstract
Background Recent studies have increasingly emphasized the strong correlation between the lipidome and the risk of pancreatic diseases. To determine causality, a Mendelian randomization (MR) analysis was performed to identify connections between the lipidome and pancreatic diseases. Methods Statistics from a genome-wide association study of the plasma lipidome, which included a diverse array of 179 lipid species, were obtained from the GeneRISK cohort study with 7,174 participants. Genetic associations with four types of pancreatitis and pancreatic cancer were sourced from the R11 release of the FinnGen consortium. Two pancreatitis datasets from UK Biobank were employed as the validation cohort. MR analysis was conducted to assess the relationship between the genetically predicted plasma lipidome and these pancreatic diseases. Inverse variance weighted was adopted as the main statistical method. Bayesian weighted MR was employed for further verification. The MR-Egger intercept test for pleiotropy and Cochrane's Q statistics test for heterogeneity were performed to ensure the robustness. Results MR analysis yielded significant evidence that 26, 25, 2, and 19 lipid species were correlated with diverse outcomes of pancreatitis, and 8 lipid species were correlated with pancreatic cancer. Notably, sterol ester (27:1/20:2) levels (OR: 0.84, 95% CI: 0.78-0.90, P = 5.79 × 10-7) were significantly associated with acute pancreatitis, and phosphatidylcholine (17:0_20:4) levels (OR: 0.89, 95% CI: 0.84-0.94, P = 1.78 × 10-4) and sterol ester (27:1/20:4) levels (OR: 0.90, 95% CI: 0.86-0.95, P = 2.71 × 10-4) levels were significantly associated with chronic pancreatitis after the Bonferroni-corrected test. As for validation, 14 and 9 lipid species were correlated with acute and chronic pancreatitis of UK Biobank. Some lipid classes showed significant effects both in the FinnGen consortium and UK Biobank datasets. Conclusions The findings of this study indicate a potential genetic predisposition linking the plasma lipidome to pancreatic diseases and good prospects for future pancreatic disease clinical trials.
Collapse
Affiliation(s)
- Liaoyi Lin
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingbao Huang
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Songzan Qian
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lifang Chen
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Houzhang Sun
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
29
|
Merrill AH. Don't Be Surprised When These Surprise You: Some Infrequently Studied Sphingoid Bases, Metabolites, and Factors That Should Be Kept in Mind During Sphingolipidomic Studies. Int J Mol Sci 2025; 26:650. [PMID: 39859363 PMCID: PMC11765627 DOI: 10.3390/ijms26020650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Sphingolipidomic mass spectrometry has provided valuable information-and surprises-about sphingolipid structures, metabolism, and functions in normal biological processes and disease. Nonetheless, many noteworthy compounds are not routinely determined, such as the following: most of the sphingoid bases that mammals biosynthesize de novo other than sphingosine (and sometimes sphinganine) or acquire from exogenous sources; infrequently considered metabolites of sphingoid bases, such as N-(methyl)n-derivatives; "ceramides" other than the most common N-acylsphingosines; and complex sphingolipids other than sphingomyelins and simple glycosphingolipids, including glucosyl- and galactosylceramides, which are usually reported as "monohexosylceramides". These and other subspecies are discussed, as well as some of the circumstances when they are likely to be seen (or present and missed) due to experimental conditions that can influence sphingolipid metabolism, uptake from the diet or from the microbiome, or as artifacts produced during extraction and analysis. If these compounds and factors are kept in mind during the design and interpretation of lipidomic studies, investigators are likely to be surprised by how often they appear and thereby advance knowledge about them.
Collapse
Affiliation(s)
- Alfred H Merrill
- School of Biological Sciences and The Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
30
|
Saeedi V, Rahimzadeh N, Ehsanipour F, Shalbaf N, Farahi A, Rashidi K, Kamalzadeh L. Clinical presentation and management challenges of sphingosine-1-phosphate lyase insufficiency syndrome associated with an SGPL1 variant: a case report. BMC Pediatr 2025; 25:1. [PMID: 39755650 DOI: 10.1186/s12887-024-05311-y] [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: 09/16/2024] [Accepted: 12/05/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND This case report describes a unique presentation of sphingosine-1-phosphate lyase insufficiency syndrome (SPLIS) caused by a rare SGPL1 variant, highlighting the diagnostic and management challenges associated with this condition. CASE PRESENTATION A 2-year-old Iranian female presented with steroid-resistant nephrotic syndrome (NS), primary adrenal insufficiency (AI), growth delay, seizures, and hyperpigmentation. Laboratory evaluation revealed hypoalbuminemia, significant proteinuria, hyperkalemia, and elevated adrenocorticotropic hormone (ACTH) levels. The patient was diagnosed with SPLIS through genetic testing, revealing a c.1018 C > T variant in SGPL1. Despite supportive treatment, including corticosteroids and cyclosporine, the patient's condition deteriorated, leading to end-stage renal disease and sepsis, ultimately resulting in death. CONCLUSIONS This case underscores the clinical heterogeneity of SPLIS and the importance of early genetic evaluation in patients with combined NS and AI. Personalized management approaches and increased awareness among clinicians are essential to improve patient outcomes.
Collapse
Affiliation(s)
- Vahid Saeedi
- Pediatric Growth and Development Research Center, Institute of Endocrinology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Nahid Rahimzadeh
- Department of Pediatrics, School of Medicine, Hazrate-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Ehsanipour
- Pediatric Growth and Development Research Center, Institute of Endocrinology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Shalbaf
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhosein Farahi
- Pediatric Growth and Development Research Center, Institute of Endocrinology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Khalil Rashidi
- Cancer Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Leila Kamalzadeh
- Geriatric Mental Health Research Center, Department of Psychiatry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Kol M, Novak AJE, Morstein J, Schröer C, Sokoya T, Mensing S, Korneev SM, Trauner D, Holthuis JCM. Optical control of sphingolipid biosynthesis using photoswitchable sphingosines. J Lipid Res 2025; 66:100724. [PMID: 39672331 PMCID: PMC11782902 DOI: 10.1016/j.jlr.2024.100724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/06/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024] Open
Abstract
Sphingolipid metabolism comprises a complex interconnected web of enzymes, metabolites, and modes of regulation that influence a wide range of cellular and physiological processes. Deciphering the biological relevance of this network is challenging as numerous intermediates of sphingolipid metabolism are short-lived molecules with often opposing biological activities. Here, we introduce clickable, azobenzene-containing sphingosines, termed caSphs, as light-sensitive substrates for sphingolipid biosynthesis. Photo-isomerization of the azobenzene moiety enables reversible switching between a straight trans- and curved cis-form of the lipid's hydrocarbon tail. Combining in vitro enzyme assays with metabolic labeling studies, we demonstrate that trans-to-cis isomerization of caSphs profoundly stimulates their metabolic conversion by ceramide synthases and downstream sphingomyelin synthases. These light-induced changes in sphingolipid production rates are acute, reversible, and can be implemented with great efficiency in living cells. Our findings establish caSphs as versatile tools for manipulating sphingolipid biosynthesis and function with the spatiotemporal precision of light.
Collapse
Affiliation(s)
- Matthijs Kol
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, Osnabrück, Germany; Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany.
| | | | - Johannes Morstein
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, USA
| | - Christian Schröer
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, Osnabrück, Germany; Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Tolulope Sokoya
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, Osnabrück, Germany; Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Svenja Mensing
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, Osnabrück, Germany; Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Sergei M Korneev
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, Osnabrück, Germany; Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Dirk Trauner
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Joost C M Holthuis
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, Osnabrück, Germany; Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany.
| |
Collapse
|
32
|
Binish F, Xiao J. Deciphering the role of sphingosine 1-phosphate in central nervous system myelination and repair. J Neurochem 2025; 169:e16228. [PMID: 39290063 DOI: 10.1111/jnc.16228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid of the sphingolipid family and plays a pivotal role in the mammalian nervous system. Indeed, S1P is a therapeutic target for treating demyelinating diseases such as multiple sclerosis. Being part of an interconnected sphingolipid metabolic network, the amount of S1P available for signalling is equilibrated between its synthetic (sphingosine kinases 1 and 2) and degradative (sphingosine 1-phosphate lyase) enzymes. Once produced, S1P exerts its biological roles via signalling to a family of five G protein-coupled S1P receptors 1-5 (S1PR1-5). Despite significant progress, the precise roles that S1P metabolism and downstream signalling play in regulating myelin formation and repair remain largely opaque and somewhat controversial. Genetic or pharmacological studies adopting various model systems identify that stimulating S1P-S1PR signalling protects myelin-forming oligodendrocytes after central nervous system (CNS) injury and attenuates demyelination in vivo. However, evidence to support its role in remyelination of the mammalian CNS is limited, although blocking S1P synthesis sheds light on the role of endogenous S1P in promoting CNS remyelination. This review focuses on summarising the current understanding of S1P in CNS myelin formation and repair, discussing the complexity of S1P-S1PR interaction and the underlying mechanism by which S1P biosynthesis and signalling regulates oligodendrocyte myelination in the healthy and injured mammalian CNS, raising new questions for future investigation.
Collapse
Affiliation(s)
- Fatima Binish
- Department of Health Sciences and Biostatistics, School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Junhua Xiao
- Department of Health Sciences and Biostatistics, School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
| |
Collapse
|
33
|
Dubot P, Sabourdy F, Levade T. Human genetic defects of sphingolipid synthesis. J Inherit Metab Dis 2025; 48:e12745. [PMID: 38706107 PMCID: PMC11730260 DOI: 10.1002/jimd.12745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024]
Abstract
Sphingolipids are ubiquitous lipids, present in the membranes of all cell types, the stratum corneum and the circulating lipoproteins. Autosomal recessive as well as dominant diseases due to disturbed sphingolipid biosynthesis have been identified, including defects in the synthesis of ceramides, sphingomyelins and glycosphingolipids. In many instances, these gene variants result in the loss of catalytic function of the mutated enzymes. Additional gene defects implicate the subcellular localization of the sphingolipid-synthesizing enzyme, the regulation of its activity, or even the function of a sphingolipid-transporter protein. The resulting metabolic alterations lead to two major, non-exclusive types of clinical manifestations: a neurological disease, more or less rapidly progressive, associated or not with intellectual disability, and an ichthyotic-type skin disorder. These phenotypes highlight the critical importance of sphingolipids in brain and skin development and homeostasis. The present article reviews the clinical symptoms, genetic and biochemical alterations, pathophysiological mechanisms and therapeutic options of this relatively novel group of metabolic diseases.
Collapse
Affiliation(s)
- Patricia Dubot
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III—Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT)ToulouseFrance
- Laboratoire de BiochimieInstitut Fédératif de Biologie, CHU PurpanToulouseFrance
- Centre de RecherchesCHU Sainte‐Justine, Université de MontréalMontréalCanada
| | - Frédérique Sabourdy
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III—Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT)ToulouseFrance
- Laboratoire de BiochimieInstitut Fédératif de Biologie, CHU PurpanToulouseFrance
| | - Thierry Levade
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III—Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT)ToulouseFrance
- Laboratoire de BiochimieInstitut Fédératif de Biologie, CHU PurpanToulouseFrance
| |
Collapse
|
34
|
Bodin S, Elhabashy H, Macdonald E, Winter D, Gauthier‐Rouvière C. Flotillins in membrane trafficking and physiopathology. Biol Cell 2025; 117:e2400134. [PMID: 39877933 PMCID: PMC11775717 DOI: 10.1111/boc.202400134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/20/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
Flotillin 1 and 2 are highly conserved and homologous members of the stomatin, prohibitin, flotillin, HflK/C (SPFH) family. These ubiquitous proteins assemble into hetero-oligomers at the cytoplasmic membrane in sphingolipid-enriched domains. Flotillins play crucial roles in various cellular processes, likely by concentrating sphingosine. They primarily act as scaffolding protein complexes within membrane microdomains (also called lipid rafts) and induce endocytosis and trafficking. Their diverse cargos in the upregulated flotillin-induced trafficking (UFIT) pathway, including tyrosine kinase receptors, adhesion molecules, and neurotransmitter receptors, link them to a wide range of cellular processes and diseases. Consequently, flotillin upregulation has been associated with various pathological conditions such as cancer, metabolic disorders, and neurodegenerative diseases. Flotillins may also be co-opted by pathogens to facilitate their entry and growth within host cells. In this review, we examined recent advancements in elucidating the structure and functions of the flotillin protein complex, including its implications in favoring the generation of sphingosine 1-phosphate, an essential bioactive lipid. We emphasized how the recent cryo-electron microscopy (cryo-EM) structure of a truncated cone-shaped cage composed of 22 copies of flotillin 1 and 2 subunits has enhanced our understanding of the flotillin complex organization within membrane microdomains and its role in membrane remodeling. We also explored how flotillin upregulation can perturb endosomal trafficking and contribute to various pathologies. A comprehensive understanding of flotillin oligomer organization and function is crucial to developing targeted therapies for diseases associated with flotillin overexpression.
Collapse
Affiliation(s)
- Stéphane Bodin
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), BIOLuMUniversity of Montpellier, CNRS UMR 5237MontpellierFrance
| | - Hadeer Elhabashy
- Department of Protein EvolutionMax Planck Institute for BiologyTübingenGermany
- Department of Computer ScienceUniversity of TübingenTübingenGermany
- Institute for Bioinformatics and Medical InformaticsUniversity of TübingenTübingenGermany
| | - Ewan Macdonald
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), BIOLuMUniversity of Montpellier, CNRS UMR 5237MontpellierFrance
| | - Dominic Winter
- Institute of Biochemistry and Molecular BiologyUniversity of BonnBonnGermany
| | - Cécile Gauthier‐Rouvière
- CRBM (Centre de Recherche en Biologie cellulaire de Montpellier), BIOLuMUniversity of Montpellier, CNRS UMR 5237MontpellierFrance
| |
Collapse
|
35
|
Rao Z, Zhang S, Xu W, Huang P, Xiao X, Hu X. Early Recognition of Secondary Asthma Caused by Lower Respiratory Tract Infection in Children Based on Multi-Omics Signature: A Retrospective Cohort Study. Int J Gen Med 2024; 17:6229-6241. [PMID: 39703797 PMCID: PMC11656193 DOI: 10.2147/ijgm.s498965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
Objective To explore the types of pathogens causing lower respiratory tract infections (LTRIs) in children and construction of a predictive model for monitoring secondary asthma caused by LTRIs. Methods Seven hundred and seventy-five children with LTRIs treated from June 2017 to July 2024 were selected as research subjects. Bacterial isolation and culture were performed on all children, and drug sensitivity tests were conducted on the isolated pathogens; And according to whether the child developed secondary asthma during treatment, they were divided into asthma group (n = 116) and non-asthma group (n = 659); Using logistic regression model to analyze the risk factors affecting secondary asthma in children with LTRIs, and establishing machine learning (ie nomogram and decision tree) prediction models; Using ROC curve analysis machine learning algorithms to predict AUC values, sensitivity, and specificity of secondary asthma in children with LTRIs. Results 792 pathogenic bacteria were isolated from 775 children with LTRIs through bacterial culture, including 261 Gram positive bacteria (32.95%) and 531 Gram negative bacteria (67.05%). Logistic regression model analysis showed that Glycerophospholipids, Sphingolipids and radiomics characteristics were risk factors for secondary asthma in children with LTRIs (P < 0.05). The AUC, sensitivity, and specificity of nomogram prediction for secondary asthma in children with LTRIs were 0.817(95CI: 0.760-0.874), 82.3%, and 76.6%, respectively; The AUC of decision tree prediction for secondary asthma in children with LTRIs is 0.926(95% CI: 0.869-0.983), with a sensitivity of 96.7% and a specificity of 87.8%. Conclusion LTRIs in children are mainly caused by Staphylococcus aureus, Streptococcus pneumoniae, Staphylococcus epidermidis, Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa; In addition, machine learning combined with multi-omics prediction models has shown good ability in predicting LTRIs combined with asthma, providing a non-invasive and effective method for clinical decision-making.
Collapse
Affiliation(s)
- Zhihui Rao
- Department of Pediatric Comprehensive Internal Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, 330008, People’s Republic of China
| | - Shuqin Zhang
- Department of Pediatric Comprehensive Internal Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, 330008, People’s Republic of China
| | - Wenlin Xu
- Department of Pediatric Comprehensive Internal Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, 330008, People’s Republic of China
| | - Pan Huang
- Department of Pediatric Comprehensive Internal Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, 330008, People’s Republic of China
| | - Xiaofei Xiao
- Department of Pediatric Comprehensive Internal Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, 330008, People’s Republic of China
| | - Xiuxiu Hu
- Department of Pediatric Comprehensive Internal Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, 330008, People’s Republic of China
| |
Collapse
|
36
|
Jackson KG, Zhao D, Su L, Lipp MK, Toler C, Idowu M, Yan Q, Wang X, Gurley E, Wu N, Puri P, Chen Q, Lesnefsky EJ, Dupree JL, Hylemon PB, Zhou H. Sphingosine kinase 2 (SphK2) depletion alters redox metabolism and enhances inflammation in a diet-induced MASH mouse model. Hepatol Commun 2024; 8:e0570. [PMID: 39773902 PMCID: PMC11567706 DOI: 10.1097/hc9.0000000000000570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/15/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Sphingosine-1 phosphate (S1P) is a bioactive lipid molecule that modulates inflammation and hepatic lipid metabolism in MASLD, which affects 1 in 3 people and increases the risk of liver fibrosis and hepatic cancer. S1P can be generated by 2 isoforms of sphingosine kinase (SphK). SphK1 is well-studied in metabolic diseases. In contrast, SphK2 function is not well characterized. Both sphingolipid and redox metabolism dysregulation contribute to MASLD pathologic progression. While SphK2 localizes to both the nucleus and mitochondria, its specific role in early MASH is not well defined. METHODS This study examined SphK2 depletion effects on hepatic redox metabolism, mitochondrial function, and inflammation in a 16-week western diet plus sugar water (WDSW)-induced mouse model of early MASH. RESULTS WDSW-SphK2-/- mice exhibit increased hepatic lipid accumulation and hepatic redox dysregulation. In addition, mitochondria-localized cholesterol and S1P precursors were increased. We traced SphK2-/--mediated mitochondrial electron transport chain impairment to respiratory complex-IV and found that decreased mitochondrial redox metabolism coincided with increased oxidase gene expression and oxylipin production. Consistent with this relationship, we observed pronounced increases in hepatic inflammatory gene expression, prostaglandin accumulation, and innate immune homing in WDSW-SphK2-/- mice compared to WDSW-wild-type mice. CONCLUSIONS These studies suggest SphK2-derived S1P maintains hepatic redox metabolism and describe the potential consequences of SphK2 depletion on proinflammatory gene expression, lipid mediator production, and immune infiltration in MASH progression.
Collapse
Affiliation(s)
- Kaitlyn G. Jackson
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Derrick Zhao
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Lianyong Su
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
- Department of Research, Richmond Veterans Healthcare System, Richmond, Virginia, USA
| | - Marissa K. Lipp
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Cameron Toler
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Michael Idowu
- Department of Pathology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Qianhua Yan
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuan Wang
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Emily Gurley
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Nan Wu
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Puneet Puri
- Department of Research, Richmond Veterans Healthcare System, Richmond, Virginia, USA
- Division of Gastroenterology, Department of Internal Medicine, Hepatology, and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Qun Chen
- Department of Internal Medicine, Cardiology, Pauley Heart Center, Richmond, Virginia, USA
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Edward J. Lesnefsky
- Department of Internal Medicine, Cardiology, Pauley Heart Center, Richmond, Virginia, USA
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jeffrey L. Dupree
- Department of Research, Richmond Veterans Healthcare System, Richmond, Virginia, USA
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Phillip B. Hylemon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
- Department of Research, Richmond Veterans Healthcare System, Richmond, Virginia, USA
| |
Collapse
|
37
|
Geng R, Guo J, Lao Y, Kang SG, Huang K, Tong T. Chronic UVB exposure induces hepatic injury in mice: Mechanistic insights from integrated multi-omics. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 362:124933. [PMID: 39265770 DOI: 10.1016/j.envpol.2024.124933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/28/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024]
Abstract
Chronic UVB exposure poses a significant threat to both skin and visceral health. In recent years, the adverse role of chronic UVB exposure in liver health has been suggested but not fully elucidated. This study aims to comprehensively investigate the effects of chronic UVB exposure on liver health in male SKH-1 hairless mice and clarify potential mechanisms through multi-omics approaches. The findings suggested that 10-week chronic skin exposure to UVB not only triggers hepatic inflammation and oxidative stress but also, more importantly, results in lipid metabolism abnormalities in the liver. Hepatic transcriptomic analysis revealed significant alterations in various signaling pathways and physiological processes associated with inflammation, oxidative stress, and lipid metabolism. Further lipidomic analysis illustrated significant changes in the metabolism of glycerolipids, sphingolipids, and glycerophospholipids in the liver following chronic UVB exposure. The 16S rRNA sequencing analysis indicated that chronic UVB exposure disrupts the structure and function of the microbiota. In search of potential mechanisms used by the microbiome to regulate the hepatic disease morphology, we filtered mouse fecal supernatants and cultured the supernatants with HepG2 cells. Fecal supernatant from UVB-exposed mice induced increased secretion of the inflammatory cytokine IL-8, accumulation of MDA, reduced SOD activity, and decreased lipid content in normal hepatic cells. In summary, skin chronic exposure to UVB induces multiple liver injuries and gut microbiota dysbiosis in mice and gut microbiota metabolites may be one of the contributing factors to hepatic injury caused by chronic UVB exposure. These discoveries deepen the comprehension of the health risks associated with chronic UVB exposure.
Collapse
Affiliation(s)
- Ruixuan Geng
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China
| | - Jingya Guo
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China
| | - Yujie Lao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China
| | - Seong-Gook Kang
- Department of Food Engineering and Solar Salt Research Center, Mokpo National University, Muangun, 58554, Republic of Korea
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China
| | - Tao Tong
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), Ministry of Agriculture, Beijing, 100083, China; Beijing Laboratory for Food Quality and Safety, Beijing, 100083, China.
| |
Collapse
|
38
|
Liu H, Li X, Liu W, Zhang C, Zhang S, Zhou X, Bode AM, Luo X. DHRS2-induced SPHK1 downregulation contributes to the cell growth inhibition by Trichothecin in colorectal carcinoma. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119846. [PMID: 39284549 DOI: 10.1016/j.bbamcr.2024.119846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/02/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Deregulation of lipid metabolism is one of the most prominent metabolic features in cancer. The activation of sphingolipid metabolic pathways affects the proliferation, invasion, angiogenesis, chemoresistance, and immune escape of tumors, including colorectal cancer (CRC). Dehydrogenase/reductase member 2 (DHRS2), which belongs to the short-chain dehydrogenase/reductase (SDR) family, has been reported to participate in the regulation of lipid metabolism and impact on cancer progression. Trichothecin (TCN) is a sesquiterpenoid metabolite originating from an endophytic fungus of the herbal plant Maytenus hookeri Loes. Studies have shown that TCN exerts a broad-spectrum antitumor activity. METHODS We evaluated the proliferative ability of CRC cells by CCK8 and colony formation assays. A metabolite profiling using liquid chromatography coupled with mass spectrometry (LC/MS) was adopted to identify the proximal metabolite changes linked to DHRS2 overexpression. RNA stability assay and RNA immunoprecipitation (RIP) experiments were applied to determine the post-transcriptional regulation of SPHK1 expression by DHRS2. We used flow cytometry to detect changes in cell cycle and cell apoptosis of CRC cells in the absence or presence of TCN. RESULTS We demonstrate that DHRS2 hampers the sphingosine kinases 1 (SPHK1)/sphingosine 1-phosphate (S1P) metabolic pathway to inhibit CRC cell growth. DHRS2 directly binds to SPHK1 mRNA to accelerate its degradation in a post-transcriptionally regulatory manner. Moreover, we illustrate that SPHK1 downregulation induced by DHRS2 contributes to TCN-induced growth inhibition of CRC. CONCLUSIONS The present study provides a mechanistic connection among metabolic enzymes, metabolites, and the malignant progression of CRC. Moreover, TCN could be developed as a potential pharmacological tool against CRC by the induction of DHRS2 and targeting SPHK1/S1P metabolic pathway.
Collapse
Affiliation(s)
- Huiwen Liu
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Xiang Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, PR China
| | - Wenbin Liu
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; Department of Pathology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Chunhong Zhang
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Shuzhao Zhang
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China
| | - Xinran Zhou
- Hengyang Medical College, University of South China, Hengyang 421001 Hunan, PR China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Xiangjian Luo
- Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China; NHC Key Laboratory of Carcinogenesis, the Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan 410078, PR China; Key Laboratory of Biological Nanotechnology of National Health Commission, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
39
|
Meacci E, Chirco A, Garcia-Gil M. Potential Vitamin E Signaling Mediators in Skeletal Muscle. Antioxidants (Basel) 2024; 13:1383. [PMID: 39594525 PMCID: PMC11591548 DOI: 10.3390/antiox13111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Vitamin E (Vit E) deficiency studies underline the relevance of this vitamin in skeletal muscle (SkM) homeostasis. The knowledge of the effectors and modulators of Vit E action in SkM cells is limited, especially in aging and chronic diseases characterized by a decline in musculoskeletal health. Vit E comprises eight fat-soluble compounds grouped into tocopherols and tocotrienols, which share the basic chemical structure but show different biological properties and potentials to prevent diseases. Vit E has antioxidant and non-antioxidant activities and both favorable and adverse effects depending on the specific conditions and tissues. In this review, we focus on the actual knowledge of Vit E forms in SkM functions and new potential signaling effectors (i.e., bioactive sphingolipids and myokines). The possible advantages of Vit E supplementation in counteracting SkM dysfunctions in sarcopenia and under microgravity will also be discussed.
Collapse
Affiliation(s)
- Elisabetta Meacci
- Department of Experimental and clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Firenze, Italy
- Interuniversity Institute of Myology, University of Florence, 50134 Firenze, Italy
| | - Antony Chirco
- Department of Experimental and clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Firenze, Italy
| | - Mercedes Garcia-Gil
- Department of Biology, Unit of Physiology, University of Pisa, Via S. Zeno 31, 56127 Pisa, Italy;
| |
Collapse
|
40
|
Feng L, He B, Xia J, Wang Z. Untargeted and Targeted Lipidomics Unveil Dynamic Lipid Metabolism Alterations in Type 2 Diabetes. Metabolites 2024; 14:610. [PMID: 39590846 PMCID: PMC11596168 DOI: 10.3390/metabo14110610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is a complex metabolic disorder with a growing body of evidence suggesting the central role of lipid metabolism in its pathogenesis. However, the dynamic changes in lipid metabolism across different stages of T2DM remain understudied. OBJECTIVE This study aimed to elucidate the temporal alterations in lipid metabolism in T2DM using an integrated lipidomics approach. METHOD Serum samples from 155 subjects were analyzed using LC-MS-based lipidomics, including untargeted and targeted approaches. RESULTS We identified significant alterations in 44 lipid metabolites in newly diagnosed T2DM patients and 29 in high-risk individuals, compared with healthy controls. Key metabolic pathways such as sphingomyelin, phosphatidylcholine, and sterol ester metabolism were disrupted, highlighting the involvement of insulin resistance and oxidative stress in T2DM progression. Moreover, 13 lipid metabolites exhibited diagnostic potential for T2DN, showing consistent trends of increase or decrease as the disease progressed. CONCLUSION Our findings underscore the importance of lipid metabolism in T2D development and identify potential lipid biomarkers for early diagnosis and monitoring of disease progression, which contribute to paving the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Li Feng
- School of Agroforestry and Medicine, The Open University of China, Beijing 100039, China;
| | - Bingshu He
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China;
| | - Jianzhen Xia
- School Hospital, Minzu University of China, Beijing 100081, China;
| | - Zhonghua Wang
- Center for Imaging and Systems Biology, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China;
| |
Collapse
|
41
|
Chen C, Zheng T, Chen Y, Li Z, Wu H. A systematic evaluation of quenching, extraction and analysis procedures for metabolomics study of the mechanism of QYSLD intervention in A549 cells. Anal Bioanal Chem 2024; 416:6621-6638. [PMID: 39467912 DOI: 10.1007/s00216-024-05563-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/08/2024] [Accepted: 09/19/2024] [Indexed: 10/30/2024]
Abstract
The preparation of cellular metabolomics samples and how to achieve comprehensive coverage of different polar metabolites in cell samples in the analysis pose a challenge for cellular metabolomics. In this study, we optimized a metabolomics protocol based on ultra-high-performance liquid chromatography high-resolution mass spectrometry (UPLC/HRMS) for the extraction and detection of metabolites in A549 cells and exploration of the intervention effect of Qi-Yu-San-Long decoction (QYSLD) on A549 cells. The results indicate that the lowest level of ATP leakage was observed when A549 cells were quenched under liquid nitrogen. MeOH/chloroform/H2O (1:2:1) extraction yielded more chromatographic peaks and excellent reproducibility, and the relative extraction efficiency of most target metabolites was also high. And we optimized the chromatographic separation conditions in both HILIC and RPLC modes, enabling comprehensive detection and analysis of metabolites with varying polarities. Then, we applied the optimized method to UPLC-Q-TOF/MS-based metabolomics of A549 cells to study the mechanism of QYSLD intervention in non-small cell lung cancer (NSCLC). The CCK-8, EdU staining, and cell cycle assay showed that QYSLD inhibited the proliferation of A549 cells by interfering with the cell cycle and blocking them in the G1 phase. A total of 36 differential metabolites associated with the antitumor effects of QYSLD on NSCLC were identified, mainly involving nicotinate and nicotinamide metabolism, sphingolipid metabolism, and glycerophospholipid metabolism. And western blotting confirmed that the change in 1-methylnicotinamide levels after QYSLD intervention was associated with the inhibition of nicotinamide N-methyltransferase expression in A549 cells.
Collapse
Affiliation(s)
- Chang Chen
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road No.103, Hefei, 230038, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula & Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of the Application and Transformation of Traditional Chinese Medicine in the Prevention and Treatment of Major Pulmonary Diseases, Hefei, 230031, China
| | - Ting Zheng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road No.103, Hefei, 230038, China
| | - Yang Chen
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zegeng Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road No.103, Hefei, 230038, China
- Anhui Province Key Laboratory of the Application and Transformation of Traditional Chinese Medicine in the Prevention and Treatment of Major Pulmonary Diseases, Hefei, 230031, China
| | - Huan Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Meishan Road No.103, Hefei, 230038, China.
- Anhui Province Key Laboratory of Chinese Medicinal Formula & Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, 230012, China.
- Anhui Province Key Laboratory of the Application and Transformation of Traditional Chinese Medicine in the Prevention and Treatment of Major Pulmonary Diseases, Hefei, 230031, China.
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
42
|
Zhang B, Zhang B, Wang T, Huang B, Cen L, Wang Z. Integrated bulk and single-cell profiling characterize sphingolipid metabolism in pancreatic cancer. BMC Cancer 2024; 24:1347. [PMID: 39487387 PMCID: PMC11531184 DOI: 10.1186/s12885-024-13114-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Abnormal sphingolipid metabolism (SM) is closely linked to the incidence of cancers. However, the role of SM in pancreatic cancer (PC) remains unclear. This study aims to explore the significance of SM in the prognosis, immune microenvironment, and treatment of PC. METHODS Single-cell and bulk transcriptome data of PC were acquired via TCGA and GEO databases. SM-related genes (SMRGs) were obtained via MSigDB database. Consensus clustering was utilized to construct SM-related molecular subtypes. LASSO and Cox regression were utilized to build SM-related prognostic signature. ESTIMATE and CIBERSORT algorithms were employed to assess the tumour immune microenvironment. OncoPredict package was used to predict drug sensitivity. CCK-8, scratch, and transwell experiments were performed to analyze the function of ANKRD22 in PC cell line PANC-1 and BxPC-3. RESULTS A total of 153 SMRGs were acquired, of which 48 were linked to PC patients' prognosis. Two SM-related subtypes (SMRGcluster A and B) were identified in PC. SMRGcluster A had a poorer outcome and more active SM process compared to SMRGcluster B. Immune analysis revealed that SMRGcluster B had higher immune and stromal scores and CD8 + T cell abundance, while SMRGcluster A had a higher tumour purity score and M0 macrophages and activated dendritic cell abundance. PC with SMRGcluster B was more susceptible to gemcitabine, paclitaxel, and oxaliplatin. Then SM-related prognostic model (including ANLN, ANKRD22, and DKK1) was built, which had a very good predictive performance. Single-cell analysis revealed that in PC microenvironment, macrophages, epithelial cells, and endothelial cells had relatively higher SM activity. ANKRD22, DKK1, and ANLN have relatively higher expression levels in epithelial cells. Cell subpopulations with high expression of ANKRD22, DKK1, and ANLN had more active SM activity. In vitro experiments showed that ANKRD22 knockdown can inhibit the proliferation, migration, and invasion of PC cells. CONCLUSION This study revealed the important significance of SM in PC and identified SM-associated molecular subtypes and prognostic model, which provided novel perspectives on the stratification, prognostic prediction, and precision treatment of PC patients.
Collapse
Affiliation(s)
- Biao Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bolin Zhang
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle- Wittenberg, University Medical Center Halle, Halle, Germany
| | - Tingxin Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Bingqian Huang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Lijun Cen
- Department of Transfusion Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China.
- Key Laboratory of Molecular Pathology in Tumors of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China.
| | - Zhizhou Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
43
|
Xu K, Shen Y, Shi L, Chen F, Zhang B, He Y, Wang Y, Liu Y, Shi G, Mi B, Zeng L, Dang S, Liu X, Yan H. Lipidomic perturbations of normal-weight adiposity phenotypes and their mediations on diet-adiposity associations. Clin Nutr 2024; 43:20-30. [PMID: 39307096 DOI: 10.1016/j.clnu.2024.09.020] [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: 02/28/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND & AIMS Normal-weight obesity (NWO) and normal-weight central obesity (NWCO) have been linked to higher cardiometabolic risks, but their etiological bases and attributable dietary factors remain unclear. In this study we therefore aimed to identify lipidomic signatures and dietary factors related to NWO and NWCO and to explore the mediation associations of lipids in diet-adiposity associations. METHODS Using a high-coverage targeted lipidomic approach, we quantified 1245 serum lipids in participants with NWO (n = 150), NWCO (n = 150), or propensity-score-matched normal-weight controls (n = 150) based on the Regional Ethnic Cohort Study in Northwest China. Consumption frequency of 28 major food items was recorded using a food frequency questionnaire. RESULTS Profound lipidomic perturbations of NWCO relative to NWO were observed, and 249 (dominantly glycerolipids) as well as 48 (dominantly glycerophospholipids) lipids were exclusively associated with NWCO or NWO. Based on strong lipidomic signatures identified by a LASSO model, phospholipid biosynthesis was the top enriched pathway of NWCO, and sphingolipid metabolism was the top pathway of NWO. Remarkably, sphingolipids were positively associated with NWO and NWCO, but lyso-phosphatidylcholines were negatively associated with them. Rice, fruit juice, and carbonated drink intakes were positively associated with the risk of NWCO. Both global and individual lipidomic signatures, including SE(28:1_22:6) and HexCer(d18:1/20:1), mediated these diet-NWCO associations (mediation proportion: 15.92%-26.10%). CONCLUSIONS Differential lipidomic signatures were identified for overall and abdominal adiposity accumulation in normal-weight individuals, underlining their core mediation roles in dietary contributions to adiposity deposition.
Collapse
Affiliation(s)
- Kun Xu
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China
| | - Yuan Shen
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, 710062, Xi' an, Shaanxi, China
| | - Fangyao Chen
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China
| | - Binyan Zhang
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China; School of Public Health, Xi'an Medical College, Xi'an, 710021, China
| | - Yafang He
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China
| | - Yutong Wang
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China
| | - Yezhou Liu
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China
| | - Guoshuai Shi
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China
| | - Baibing Mi
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China
| | - Lingxia Zeng
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China
| | - Shaonong Dang
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China.
| | - Xin Liu
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China.
| | - Hong Yan
- Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China; Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Xi'an Jiaotong University, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China; Nutrition and Food Safety Engineering Research Center of Shaanxi Province, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, 76 West Yanta Road, 710061, Xi'an, Shaanxi, China.
| |
Collapse
|
44
|
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
|
45
|
Limbu KR, Chhetri RB, Kim S, Shrestha J, Oh YS, Baek DJ, Park EY. Targeting sphingosine 1-phosphate and sphingosine kinases in pancreatic cancer: mechanisms and therapeutic potential. Cancer Cell Int 2024; 24:353. [PMID: 39462385 PMCID: PMC11514880 DOI: 10.1186/s12935-024-03535-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Pancreatic cancer is known to be the most lethal cancer. Fewer new treatments are being developed for pancreatic cancer as compared to other cancers. The bioactive lipid S1P, which is mainly regulated by sphingosine kinase 1 (SK1) and sphingosine kinase 2 (SK2) enzymes, plays significant roles in pancreatic cancer initiation and exacerbation. S1P controls many signaling pathways to modulate the progression of pancreatic cancer through the G-coupled receptor S1PR1-5. Several papers reporting amelioration of pancreatic cancer via modulation of S1P levels or downstream signaling pathways have previously been published. In this paper, for the first time, we have reviewed the results of previous studies to understand how S1P and its receptors contribute to the development of pancreatic cancer, and whether S1P can be a therapeutic target. In addition, we have also reviewed papers dealing with the effects of SK1 and SK2, which are kinases that regulate the level of S1P, on the pathogenesis of pancreatic cancer. We have also listed available drugs that particularly focus on S1P, S1PRs, SK1, and SK2 for the treatment of pancreatic cancer. Through this review, we would like to suggest that the SK/S1P/S1PR signaling system can be an important target for treating pancreatic cancer, where a new treatment target is desperately warranted.
Collapse
Affiliation(s)
- Khem Raj Limbu
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | | | - Subin Kim
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | - Jitendra Shrestha
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam, 13135, South Korea
| | - Dong Jae Baek
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| |
Collapse
|
46
|
Fu F, Li W, Zheng X, Wu Y, Du D, Han C. Role of Sphingosine-1-Phosphate Signaling Pathway in Pancreatic Diseases. Int J Mol Sci 2024; 25:11474. [PMID: 39519028 PMCID: PMC11545938 DOI: 10.3390/ijms252111474] [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: 09/30/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Sphingosine-1-phosphate (S1P) is a sphingolipid metabolic product produced via the phosphorylation of sphingosine by sphingosine kinases (SPHKs), serving as a powerful modulator of various cellular processes through its interaction with S1P receptors (S1PRs). Currently, this incompletely understood mechanism in pancreatic diseases including pancreatitis and pancreatic cancer, largely limits therapeutic options for these disorders. Recent evidence indicates that S1P significantly contributes to pancreatic diseases by modulating inflammation, promoting pyroptosis in pancreatic acinar cells, regulating the activation of pancreatic stellate cells, and affecting organelle functions in pancreatic cancer cells. Nevertheless, no review has encapsulated these advancements. Thus, this review compiles information about the involvement of S1P signaling in exocrine pancreatic disorders, including acute pancreatitis, chronic pancreatitis, and pancreatic cancer, as well as prospective treatment strategies to target S1P signaling for these conditions. The insights presented here possess the potential to offer valuable guidance for the implementation of therapies targeting S1P signaling in various pancreatic diseases.
Collapse
Affiliation(s)
- Fei Fu
- West China Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610213, China; (W.L.); (X.Z.); (Y.W.)
| | - Wanmeng Li
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610213, China; (W.L.); (X.Z.); (Y.W.)
| | - Xiaoyin Zheng
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610213, China; (W.L.); (X.Z.); (Y.W.)
| | - Yaling Wu
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610213, China; (W.L.); (X.Z.); (Y.W.)
| | - Dan Du
- West China Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610213, China; (W.L.); (X.Z.); (Y.W.)
| | - Chenxia Han
- West China Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
| |
Collapse
|
47
|
Kol M, Novak AJ, Morstein J, Schröer C, Sokoya T, Mensing S, Korneev SM, Trauner D, Holthuis JC. Optical control of sphingolipid biosynthesis using photoswitchable sphingosines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.619506. [PMID: 39484495 PMCID: PMC11527141 DOI: 10.1101/2024.10.24.619506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Sphingolipid metabolism comprises a complex interconnected web of enzymes, metabolites and modes of regulation that influence a wide range of cellular and physiological processes. Deciphering the biological relevance of this network is challenging as numerous intermediates of sphingolipid metabolism are short-lived molecules with often opposing biological activities. Here, we introduce clickable, azobenzene-containing sphingosines, termed caSphs, as light-sensitive substrates for sphingolipid biosynthesis. Photo-isomerization of the azobenzene moiety enables reversible switching between a straight trans- and curved cis-form of the lipid's hydrocarbon tail. Combining in vitro enzyme assays with metabolic labeling studies, we demonstrate that trans-to-cis isomerization of caSphs profoundly stimulates their metabolic conversion by ceramide synthases and downstream sphingomyelin synthases. These light-induced changes in sphingolipid production rates are acute, reversible, and can be implemented with great efficiency in living cells. Our findings establish caSphs as versatile tools with unprecedented opportunities to manipulate sphingolipid biosynthesis and function with the spatiotemporal precision of light.
Collapse
Affiliation(s)
- Matthijs Kol
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
- Center for Cellular Nanoanalytics, Osnabrück University, Artilleriestraße 77, 49076 Osnabrück, Germany
| | - Alexander J.E. Novak
- Department of Chemistry, New York University 100 Washington Square East, New York, NY, 10003, USA
| | - Johannes Morstein
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Christian Schröer
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
- Center for Cellular Nanoanalytics, Osnabrück University, Artilleriestraße 77, 49076 Osnabrück, Germany
| | - Tolulope Sokoya
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
- Center for Cellular Nanoanalytics, Osnabrück University, Artilleriestraße 77, 49076 Osnabrück, Germany
| | - Svenja Mensing
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
- Center for Cellular Nanoanalytics, Osnabrück University, Artilleriestraße 77, 49076 Osnabrück, Germany
| | - Sergei M. Korneev
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
- Center for Cellular Nanoanalytics, Osnabrück University, Artilleriestraße 77, 49076 Osnabrück, Germany
| | - Dirk Trauner
- Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, PA, 19104, USA
| | - Joost C.M. Holthuis
- Molecular Cell Biology Division, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
- Center for Cellular Nanoanalytics, Osnabrück University, Artilleriestraße 77, 49076 Osnabrück, Germany
| |
Collapse
|
48
|
Petrovic S, Mouskeftara T, Paunovic M, Deda O, Vucic V, Milosevic M, Gika H. Unveiling Lipidomic Alterations in Metabolic Syndrome: A Study of Plasma, Liver, and Adipose Tissues in a Dietary-Induced Rat Model. Nutrients 2024; 16:3466. [PMID: 39458462 PMCID: PMC11509917 DOI: 10.3390/nu16203466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic syndrome (MetS) is a complex condition characterized by fat accumulation, dyslipidemia, impaired glucose control and hypertension. In this study, rats were fed a high-fat high-fructose (HFF) diet in order to develop MetS. After ten weeks, the dietary-induced MetS was confirmed by higher body fat percentage, lower HDL-cholesterol and increased blood pressure in the HFF-fed rats compared to the normal-fed control animals. However, the effect of MetS development on the lipidomic signature of the dietary-challenged rats remains to be investigated. To reveal the contribution of specific lipids to the development of MetS, the lipid profiling of rat tissues particularly susceptible to MetS was performed using untargeted UHPLC-QTOF-MS/MS lipidomic analysis. A total of 37 lipid species (mainly phospholipids, triglycerides, sphingolipids, cholesterol esters, and diglycerides) in plasma, 43 lipid species in liver, and 11 lipid species in adipose tissue were identified as dysregulated between the control and MetS groups. Changes in the lipid signature of selected tissues additionally revealed systemic changes in the dietary-induced rat model of MetS.
Collapse
Affiliation(s)
- Snjezana Petrovic
- Group for Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.P.); (V.V.)
| | - Thomai Mouskeftara
- Laboratory of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Marija Paunovic
- Group for Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.P.); (V.V.)
| | - Olga Deda
- Laboratory of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Biomic AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center B1.4, 10th km Thessaloniki-Thermi Rd, P.O. Box 8318, 57001 Thessaloniki, Greece
| | - Vesna Vucic
- Group for Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (M.P.); (V.V.)
| | - Maja Milosevic
- Group for Neuroendocrinology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Helen Gika
- Laboratory of Forensic Medicine & Toxicology, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Biomic AUTh, Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Balkan Center B1.4, 10th km Thessaloniki-Thermi Rd, P.O. Box 8318, 57001 Thessaloniki, Greece
| |
Collapse
|
49
|
Wang Q, Chen L, Zhang J, Liu Y, Jin Y, Wu J, Ren Z. High temperature ameliorates high-fat diet-induced obesity by promoting ceramide breakdown in skeletal muscle tissue. LIFE METABOLISM 2024; 3:loae012. [PMID: 39872144 PMCID: PMC11749596 DOI: 10.1093/lifemeta/loae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 03/03/2024] [Accepted: 04/02/2024] [Indexed: 01/29/2025]
Abstract
Obesity is considered an epidemic often accompanied by insulin resistance (IR). Heat treatment (HT) has been shown to prevent high-fat diet-induced IR in skeletal muscle, but the underlying mechanisms are poorly understood. In this study, we discovered that high temperature alleviated the hallmarks of obesity by promoting glycogen synthesis and lowering blood glucose levels in skeletal muscle tissue (SMT). Additionally, HT maintained the decay phase of heat shock factor 1 (HSF1), leading to the activation of gene expression of heat shock proteins (HSPs), which contributed to the alleviation of IR in SMT of diet-induced obese (DIO) mice. Metabolomics and lipidomics analyses showed that HT promoted ceramide (Cer) breakdown, resulting in an elevation of both sphingomyelin (SM) and sphingosine, which further contributed to the amelioration of IR in SMT of DIO mice. Importantly, the increase in sphingosine was attributed to the heightened expression of the acid ceramidase N-acylsphingosine amidohydrolase 1 (ASAH1), and the inhibition of ASAH1 attenuated HT-relieved IR in SMT of DIO mice. Surprisingly, high temperature increased the composition of Cer and cholesteryl ester in lipid droplets of skeletal muscle cells. This not only helped alleviate IR but also prevented lipotoxicity in SMT of DIO mice. These findings revealed a previously unknown connection between a high-temperature environment and sphingolipid metabolism in obesity, suggesting that high temperature can improve IR by promoting Cer catabolism in SMT of obese mice.
Collapse
Affiliation(s)
- Qiankun Wang
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Lupeng Chen
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Junzhi Zhang
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yue Liu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yi Jin
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei 430070, China
- Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China
| |
Collapse
|
50
|
Prell A, Wigger D, Huwiler A, Schumacher F, Kleuser B. The sphingosine kinase 2 inhibitors ABC294640 and K145 elevate (dihydro)sphingosine 1-phosphate levels in various cells. J Lipid Res 2024; 65:100631. [PMID: 39182604 PMCID: PMC11465068 DOI: 10.1016/j.jlr.2024.100631] [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: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
Sphingosine kinases (SphKs), enzymes that produce the bioactive lipids dihydrosphingosine 1-phosphate (dhS1P) and sphingosine 1-phosphate (S1P), are associated with various diseases, including cancer and infections. For this reason, a number of SphK inhibitors have been developed. Although off-target effects have been described for selected agents, SphK inhibitors are mostly used in research without monitoring the effects on the sphingolipidome. We have now investigated the effects of seven commonly used SphK inhibitors (5c, ABC294640 (opaganib), N,N-dimethylsphingosine, K145, PF-543, SLM6031434, and SKI-II) on profiles of selected sphingolipids in Chang, HepG2, and human umbilical vein endothelial cells. While we observed the expected (dh)S1P reduction for N,N-dimethylsphingosine, PF-543, SKI-II, and SLM6031434, 5c showed hardly any effect. Remarkably, for K145 and ABC294640, both reported to be specific for SphK2, we observed dose-dependent strong increases in dhS1P and S1P across cell lines. Compensatory effects of SphK1 could be excluded, as this observation was also made in SphK1-deficient HK-2 cells. Furthermore, we observed effects on dihydroceramide desaturase activity for all inhibitors tested, as has been previously noted for ABC294640 and SKI-II. In additional mechanistic studies, we investigated the massive increase of dhS1P and S1P after short-term cell treatment with ABC294640 and K145 in more detail. We found that both compounds affect sphingolipid de novo synthesis, with 3-ketodihydrosphingosine reductase and dihydroceramide desaturase as their targets. Our study indicates that none of the seven SphK inhibitors tested was free of unexpected on-target and/or off-target effects. Therefore, it is important to monitor cellular sphingolipid profiles when SphK inhibitors are used in mechanistic studies.
Collapse
Affiliation(s)
- Agata Prell
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Dominik Wigger
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Andrea Huwiler
- Institute of Pharmacology, Inselspital, INO-F, University of Bern, Bern, Switzerland
| | - Fabian Schumacher
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Burkhard Kleuser
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|