1
|
van Kruining D, Losen M, Dehairs J, Swinnen JV, Waelkens E, Honing M, Martinez-Martinez P. Early plasma ceramide and sphingomyelin levels reflect APOE genotype but not familial Alzheimer's disease gene mutations in female 5xFAD mice, with brain-region specific sphingolipid alterations. Neurobiol Dis 2025; 210:106923. [PMID: 40253012 DOI: 10.1016/j.nbd.2025.106923] [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: 02/28/2025] [Revised: 04/16/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025] Open
Abstract
Pathophysiological changes associated with Alzheimer's disease (AD) begin decades before dementia onset, with age and APOE ε4 genotype as major risk factors [1-4]. Primary risk factors for developing AD include aging and number of copies of the apolipoprotein E (APOE) ε4 allele. Altered sphingolipid metabolism is increasingly implicated in early AD. However, the relationship between early plasma and brain sphingolipid changes-particularly in the context of APOE genotype-remains poorly defined. In this study, we analyzed plasma and brain sphingolipid profiles in transgenic AD mice carrying human APOE3 or APOE4 variants, with or without familial AD mutations (E3FAD and E4FAD). Using liquid chromatography-tandem mass spectrometry (LC-MS/MS), we assessed 110 sphingolipid species across four major classes (ceramides (Cers), hexosylceramides (HexCers), lactosylceramides (LacCers), and sphingomyelins (SMs)) at 2, 4, and 6 months in plasma and at 6 months in brain tissue in the cortex, hippocampus, striatum, and cerebellum. Our results demonstrate that early plasma sphingolipid alterations are largely driven by APOE genotype rather than AD pathology. Specifically, APOE4 carriers showed significant increases in SM species and reductions in Cer species compared to APOE3 carriers, independent of age or AD genotype. Brain lipid profiles showed minimal changes across genotypes after region correction. However, combined p-value analyses revealed APOE- and EFAD-dependent differences in the composition of primarily cortical sphingolipids. ROC analyses demonstrated high discriminative power of plasma sphingolipids for APOE, but not for AD genotype. These findings suggest that early plasma lipid profiles in female 5xFAD mice are more strongly influenced by APOE genotype than by overt AD pathology, potentially reflecting systemic pathways linked to APOE4-associated AD risk, while early disease-associated changes in the brain appear to be subtle and region-specific. These results underscore the importance of accounting for APOE genotype in early-stage AD lipidomic studies and in the interpretation of peripheral lipid biomarkers.
Collapse
Affiliation(s)
- Daan van Kruining
- School for Mental Health and Neuroscience, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Pharmacology, University of Oxford, Oxford, UK.
| | - Mario Losen
- School for Mental Health and Neuroscience, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Pharmacology, University of Oxford, Oxford, UK
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, KU Leuven, Leuven 3000, Belgium
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, KU Leuven, Leuven 3000, Belgium
| | - Etienne Waelkens
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Leuven 3000, Belgium
| | - Maarten Honing
- Maastricht Multimodal Molecular Imaging Institute (M4I), University of Maastricht, the Netherlands
| | - Pilar Martinez-Martinez
- School for Mental Health and Neuroscience, Faculty of Health, Medicine, and Life Sciences, Maastricht University, Maastricht, the Netherlands; Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
2
|
Hu W, Bei Y, Chen G, Xu J, Yang M, Yu L, He W, Hu Y, Mao F, Chen S, Xu D, Dai H. Causal insights into the role of metabolites in venous thromboembolism pathogenesis: a metabolome-wide Mendelian randomization study. J Thromb Haemost 2025; 23:1953-1967. [PMID: 40157505 DOI: 10.1016/j.jtha.2025.03.022] [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: 12/02/2024] [Revised: 02/06/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Venous thromboembolism (VTE) is a significant global health burden, and metabolic alterations play a key role in its pathogenesis. However, previous studies have been constrained by several limitations, hindering clarification of the causal role of metabolites. OBJECTIVES To comprehensively evaluate the causal roles of metabolites in the pathogenesis of VTE and determine whether metabolites mediate the relationships between modifiable risk factors and VTE. METHODS Genetic associations involving 690 plasmas and 211 urinary metabolites were analyzed as exposures, while the outcomes for VTE were derived from a large-scale meta-analysis of genome-wide association studies. Metabolome-wide Mendelian randomization and colocalization analyses were performed to assess the causal role of metabolites in VTE. Metabolic pathway analysis was performed using MetOrigin, and druggability assessments were conducted to prioritize potential therapeutic targets. Additionally, a 2-step Mendelian randomization framework was employed to elucidate the mediating effects of metabolites on the relationships between modifiable risk factors and VTE. RESULTS After Bonferroni correction, 51 plasma metabolites and 18 urinary metabolites were significantly associated with VTE risk. Colocalization evidence supported causal relationships for 37 metabolites with VTE. Eleven metabolic pathways were identified for VTE-related metabolites, and 6 metabolites were prioritized as potential therapeutic targets. Twenty-four modifiable risk factors were associated with 28 VTE-related metabolites, 7 of which were linked to VTE risk. Mediation analyses further revealed significant mediating effect of 8 metabolites on how 6 modifiable factors influenced VTE. CONCLUSION This study identifies potential metabolite biomarkers associated with VTE risk and uncovered the metabolic mediators between modifiable risk factors and VTE, offering new insights for future prevention and treatment strategies.
Collapse
Affiliation(s)
- Wei Hu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Bei
- Department of Pharmacy, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Guoquan Chen
- Department of Pharmacy, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Junjun Xu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingdong Yang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingyan Yu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei He
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yani Hu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fengqian Mao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shunan Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Donghang Xu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haibin Dai
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; Research Center for Clinical Pharmacy, Zhejiang University, Hangzhou, China.
| |
Collapse
|
3
|
Obafemi BA, Adedara IA, Delgado CP, Obafemi OT, Aschner M, Rocha JB. Fumonisin B1 neurotoxicity: Preclinical evidence, biochemical mechanisms and therapeutic strategies. Toxicol Rep 2025; 14:101931. [PMID: 39980663 PMCID: PMC11841125 DOI: 10.1016/j.toxrep.2025.101931] [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: 12/05/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/22/2025] Open
Abstract
The neurotoxic effects of fungal toxins in both humans and animals have been well documented. Fumonisin B1 (FB1), a mycotoxin produced by fungi of the Fusarium species, is the most toxic fumonisin variant whose neurotoxic effect is still being elucidated. This review highlights the biochemical aspects of FB1 neurotoxicity, such as its mechanisms of action as well as therapeutic strategies. Both in vitro and in vivo studies have demonstrated that alteration in sphingolipid metabolism is a major event in FB-induced neurotoxicity. Studies have also shown that neurotoxicity due to FB1 involves dysregulation of several biochemical events in the brain, such as induction of oxidative stress and inflammation, mitochondrial dysfunction and associated programmed cell death, inhibition of acetylcholinesterase and alteration of neurotransmitter levels, decreased activity of Na+K+ ATPase, as well as disruption of blood-brain barrier. This review highlights the potential public health effects of FB1-induced neurotoxicity and the need to limit human and animal exposure to FB1in order to prevent its neurotoxic effect. Moreover, it is hoped that this review would stimulate studies aimed at filling the current research gaps such as delineating the effect of FB1 on the blood-brain barrier and appropriate therapies for neurotoxicity caused by FB1.
Collapse
Affiliation(s)
- Blessing A. Obafemi
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, Camobi, Santa Maria 97105-900, Brazil
- Department of Medical Biochemistry, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Isaac A. Adedara
- Department of Food Science and Technology, Center of Rural Sciences, Federal University of Santa Maria, Camobi, Santa Maria, RS 97105-900, Brazil
| | - Cássia Pereira Delgado
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, Camobi, Santa Maria 97105-900, Brazil
| | - Olabisi T. Obafemi
- Department of Life and Consumer Sciences, University of South Africa, Florida 1710 Johannesburg, South Africa
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Joao B.T. Rocha
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, Camobi, Santa Maria 97105-900, Brazil
| |
Collapse
|
4
|
Wang Y, Wu H, Hu X. Quantification of the inputs and outputs of serine and glycine metabolism in cancer cells. Arch Biochem Biophys 2025; 768:110367. [PMID: 40032043 DOI: 10.1016/j.abb.2025.110367] [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] [Revised: 10/10/2024] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND The significance of serine and glycine metabolism in cancer cells is increasingly acknowledged, yet the quantification of their metabolic flux remains incomplete, impeding a comprehensive understanding. This study aimed to quantify the metabolic flux of serine and glycine in cancer cells, focusing on their inputs and outputs, by means of Combinations of C-13 Isotopes Tracing and mathematical delineation, alongside Isotopically Nonstationary Metabolic Flux Analysis. RESULTS In HeLa cells, serine uptake, the serine synthesis pathway (SSP), and other sources (e.g., protein degradation) contribute 71.2 %, 24.0 %, and 5.7 %, respectively, to serine inputs. Conversely, glycine inputs stem from uptake (45.6 %), conversion from serine (45.1 %), and other sources (9.4 %). Serine input flux surpasses glycine by 7.3-fold. Serine predominantly directs a major fraction (94.7 %) to phospholipid, sphingolipid, and protein synthesis, with only a minor fraction (5.3 %) directing towards one-carbon unit and glycine production. Glycine mainly supports protein and nucleotide synthesis (100 %), without conversion back to serine. Serine output rate exceeds glycine output rate by 7.3-fold. Serine deprivation mainly impairs output to synthesis of phospholipid and sphingolipid, crucial for cell growth, while other outputs unaffected. AGS cells exhibit comparable serine and glycine flux to HeLa cells, albeit lacking SSP activity. Serine deprivation in AGS cells halts output flux to phospholipid, sphingolipid, protein synthesis, completely inhibiting cell growth. CONCLUSIONS By providing quantitative insights into serine and glycine metabolism, this study delineates the association of serine flux to different metabolic pathway with cancer cell growth and offers potential targets for therapeutic intervention, highlighting the importance of serine flux to pathway for the synthesis of phospholipids and sphingolipids in cancer cells growth.
Collapse
Affiliation(s)
- Yuqi Wang
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, China; Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Hao Wu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Xun Hu
- Cancer Institute (Key Laboratory for Cancer Intervention and Prevention, China National Ministry of Education, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, China.
| |
Collapse
|
5
|
Uranbileg B, Hoshino Y, Ezaka M, Kurano M, Uchida K, Yatomi Y, Ito N. Metabolism of sphingolipids in a rat spinal cord stenosis model. Biochem Biophys Rep 2025; 42:102025. [PMID: 40342530 PMCID: PMC12059668 DOI: 10.1016/j.bbrep.2025.102025] [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: 11/25/2024] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 05/11/2025] Open
Abstract
Background Lumbar spinal canal stenosis (LSCS) plays a crucial role in neurogenic claudication and neuropathic pain. Recent studies suggest that changes in sphingolipid metabolism are linked to neuropathic pain. To explore the association between sphingolipids and LSCS, we measured the levels of sphingolipids and sphingolipid-associated molecules in an animal model of cauda equina compression (CEC), a typical type of LSCS. Methods By placing silicon blocks within the lumbar epidural space, CEC model were constructed in which motor disfunction had already been confirmed in our previous study. Quantitative measurements of various sphingolipids were conducted using LC-MS/MS in spinal cord and cerebrospinal fluid (CSF) samples on days 1, 7, and 28 following insertion of silicon blocks. Additionally, gene expression was analyzed in spinal cord tissue. Results In the CEC model, there was a significant increase ceramide levels in the CSF with upregulation of ceramide synthase 1 in the spinal cord tissue samples on day 1. Further, S1P levels in the CSF increased on day 7 and in the spinal cord significantly increased on day 28, and there was an increase in mRNA expression levels of sphingosine kinases (SphK)1 on days 1,7, and 28, while SphK2 on days 7 and 28. Regarding S1P receptors, there was an increase in mRNA expression levels of S1P1 on days 1,7, and 28 and S1P3 on day1. Conclusion The production and activation of the sphingolipid signaling pathway could play a pivotal role in neuropathic pain related to LSCS.
Collapse
Affiliation(s)
- Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoko Hoshino
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Mariko Ezaka
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kanji Uchida
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuko Ito
- Department of Anesthesiology and Pain Relief Center, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
6
|
Moldovan A, Wagner F, Schumacher F, Wigger D, Kessie DK, Rühling M, Stelzner K, Tschertok R, Kersting L, Fink J, Seibel J, Kleuser B, Rudel T. Chlamydia trachomatis exploits sphingolipid metabolic pathways during infection of phagocytes. mBio 2025; 16:e0398124. [PMID: 40249190 PMCID: PMC12077188 DOI: 10.1128/mbio.03981-24] [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: 01/13/2025] [Accepted: 02/19/2025] [Indexed: 04/19/2025] Open
Abstract
Chlamydiae are obligate intracellular pathogens that utilize host cell metabolites for catabolic and anabolic processes. The bacteria replicate in epithelial cells from which they take up sphingolipids (SL) and incorporate them into the chlamydial membrane and the vacuole (termed inclusion). SL uptake is essential for Chlamydia trachomatis (Ctr) in epithelial cells; however, they can also infect phagocytes, but the consequences for the SL metabolism have not yet been investigated in these cells. We performed a quantitative sphingolipidome analysis of infected primary neutrophils, macrophages, and immortalized fallopian tube epithelial cells. Sphingosine (Sph) levels are elevated in primary M2-like macrophages and human neutrophils infected with C. trachomatis. Human neutrophils respond to the pathogen by markedly upregulating sphingosine kinase 1 (SPHK1). We show in M2-like macrophages, by RNAseq, that two counteracting pathways involving upregulation of SPHK1, but also sphingosine-1-phosphate phosphatases 1 and 2 (SGPP1 and SGPP2) and sphingosine-1-phosphate lyase (SGPL1), maintain a steady pool of S1P. Using click chemistry, we show that exogenously added sphingomyelin (SM) and ceramide (Cer) are efficiently taken up into the chlamydial inclusion and are integrated into bacterial membranes in infected M2-like macrophages. Exogenous Sph reduces chlamydial infectivity, is transported into the inclusion lumen, and integrates into chlamydial membranes, suggesting that this particular SL species could represent a host defense mechanism. Taken together, our data indicate an important role for Sph/Sph kinase vs S1P/S1P phosphatase balance in infected phagocytes and a previously unrecognized role for sphingosine in the immune defense against chlamydial infection.IMPORTANCEChlamydia trachomatis (Ctr) is the leading cause of sexually transmitted diseases worldwide. Left untreated, it can cause severe complications such as blindness, pelvic inflammatory disease, or infertility. To date, no vaccines are available, and antibiotic treatment represents the only therapeutic approach to cure the infection. Limited access to antibiotics and displaced antibiotic intake increase the risk of developing recurring infections. Immune cells which fail to clear the infection and serve as a niche for chlamydial survival and replication, favor this outcome. Our research aims to elucidate the influence of sphingolipids (SL) during chlamydial infection, especially of phagocytic cells. Identifying relevant targets offers new strategies to develop alternative treatment methods.
Collapse
Affiliation(s)
- Adriana Moldovan
- Department of Microbiology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Fabienne Wagner
- Department of Microbiology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Fabian Schumacher
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Berlin, Germany
| | - Dominik Wigger
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Berlin, Germany
| | - David Komla Kessie
- Department of Microbiology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Marcel Rühling
- Department of Microbiology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Kathrin Stelzner
- Department of Microbiology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Regina Tschertok
- Department of Microbiology, University of Würzburg, Würzburg, Bavaria, Germany
| | - Louise Kersting
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Bavaria, Germany
| | - Julian Fink
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Bavaria, Germany
| | - Jürgen Seibel
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Bavaria, Germany
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Berlin, Germany
| | - Thomas Rudel
- Department of Microbiology, University of Würzburg, Würzburg, Bavaria, Germany
| |
Collapse
|
7
|
Mondal K, Gary AA, Dash A, Del Mar NA, Stephenson DJ, Chalfant CE, Reiner A, Sears B, Mandal N. Oral Supplementation of n-3 Polyunsaturated Fatty Acids (n-3-PUFA) Can Prevent TBI-Induced Visual, Motor, and Emotional Deficits in Mice. Mol Neurobiol 2025:10.1007/s12035-025-05019-9. [PMID: 40346443 DOI: 10.1007/s12035-025-05019-9] [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: 02/20/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
Traumatic brain injury (TBI) causes neuroinflammation and can generate long-term pathological consequences, including motor and visual impairments, cognitive deficits, and depression. In our previous study, we found that Fat1+-transgenic mice with higher endogenous n-3 polyunsaturated fatty acids (n-3 PUFA) were protected from post-TBI behavioral deficits and exhibited reduced levels of TBI-induced microglial activation, inflammatory factors, and sphingolipid ceramide, a lipid mediator of inflammation and cell death. This study's objective was to evaluate if feeding n-3 PUFA (EPA and docosahexaenoic acid, DHA 2:1) could restrict the elevation of ceramide in brain tissue and prevent TBI-mediated sensory-motor and behavioral deficits. Wildtype C57/BL6 mice were gavage pre-fed with PUFA (EPA: DHA = 2:1) at 500 mg/kg body weight/week for 2 weeks before and 4 weeks after exposure to left side focal cranial air-blast (50 psi) TBI or sham-blast (0-psi). Saline-gavaged mice served as controls. Following blast injury, various motor, visual, and behavioral tests were conducted, and brain tissues were collected for histological and biochemical assays. Lipidomics analysis confirmed a significant elevation of EPA in the plasma and brain tissue of PUFA-fed mice. TBI-Blast brain tissues were found to have elevated ceramide levels in control mice but not in PUFA-fed mice. Moreover, PUFA-fed mice demonstrated protection against motor impairment, photoreceptor dysfunction, depression, oculomotor nerve degeneration, and microglia activation in the optic tract. Our results demonstrate that EPA-mediated suppression of ceramide biosynthesis and neuroinflammatory factors in PUFA-fed mice is associated with significant protection against the visual, motor, and emotional deficits caused by TBI.
Collapse
Affiliation(s)
- Koushik Mondal
- Department of Ophthalmology, The University of Health Science Centre, 930 Madison Ave., Suite 718, Memphis, TN, 38163, USA
- Molecular Diagnostics Laboratory, Department of Basic & Translational Research, Saroj Gupta Cancer Centre & Research Institute, Kolkata, WB, 700 063, India
| | - Ashlyn A Gary
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Anisha Dash
- ETSU Quillen College of Medicine, Mountain Home, Johnson City, TN, 37684, USA
| | - Nobel A Del Mar
- Department of Ophthalmology, The University of Health Science Centre, 930 Madison Ave., Suite 718, Memphis, TN, 38163, USA
| | - Daniel J Stephenson
- Departments of Medicine and Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Charles E Chalfant
- Departments of Medicine and Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
- Research Service, Richmond VA Medical Center, Richmond, VA, 23298, USA
| | - Anton Reiner
- Department of Ophthalmology, The University of Health Science Centre, 930 Madison Ave., Suite 718, Memphis, TN, 38163, USA
- Department of Anatomy and Neurobiology, The University of Health Science Centre, Memphis, TN, 38163, USA
| | - Barry Sears
- Inflammation Research Foundation, Peabody, MA, 01960, USA
| | - Nawajes Mandal
- Department of Ophthalmology, The University of Health Science Centre, 930 Madison Ave., Suite 718, Memphis, TN, 38163, USA.
- Department of Anatomy and Neurobiology, The University of Health Science Centre, Memphis, TN, 38163, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Health Science Centre, Memphis, TN, 38163, USA.
- Memphis VA Medical Center, Memphis, TN, 38104, USA.
| |
Collapse
|
8
|
Zhang R, Vooijs MA, Keulers TG. Key Mechanisms in Lysosome Stability, Degradation and Repair. Mol Cell Biol 2025; 45:212-224. [PMID: 40340648 DOI: 10.1080/10985549.2025.2494762] [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: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 05/10/2025] Open
Abstract
Lysosomes are organelles that play pivotal roles in macromolecule digestion, signal transduction, autophagy, and cellular homeostasis. Lysosome instability, including the inhibition of lysosomal intracellular activity and the leakage of their contents, is associated with various pathologies, including cancer, neurodegenerative diseases, inflammatory diseases and infections. These lysosomal-related pathologies highlight the significance of factors contributing to lysosomal dysfunction. The vulnerability of the lysosomal membrane and its components to internal and external stimuli make lysosomes particularly susceptible to damage. Cells are equipped with mechanisms to repair or degrade damaged lysosomes to prevent cell death. Understanding the factors influencing lysosome stabilization and damage repair is essential for developing effective therapeutic interventions for diseases. This review explores the factors affecting lysosome acidification, membrane integrity, and functional homeostasis and examines the underlying mechanisms of lysosomal damage repair. In addition, we summarize how various risk factors impact lysosomal activity and cell fate.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Radiation Oncology (MAASTRO)/GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Marc A Vooijs
- Department of Radiation Oncology (MAASTRO)/GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Tom Gh Keulers
- Department of Radiation Oncology (MAASTRO)/GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
9
|
Ghallab DS, Shawky E, Khalifa AA, Elblehi SS, Mohyeldin MM, Ibrahim RS. Unveiling the pharmacological mechanisms of Spirulina platensis in rheumatoid arthritis rats through the integration of serum metabolomics, pathways analysis, and experimental validation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04191-y. [PMID: 40332553 DOI: 10.1007/s00210-025-04191-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease primarily manifested by insistent proliferative synovitis, joint degradation, and bone erosions with no targeted therapy yet. Spirulina platensis serves as a treasure house of bioactive compounds with potential significance against different inflammatory ailments. Inspired by the potentiating biological attributes of S. platensis, the current investigation is concerned with dissecting the mechanistic basis of S. platensis against rheumatoid arthritis (RA) through a series of biochemical and histopathological assessments integrated with a serum metabolomics strategy to explore more efficacious and safe alternative therapies to rectify RA. Firstly, a rat model of RA was established using complete Freund's adjuvant (CFA), and RA-related biochemical and histopathological scores were determined as monitoring indexes for control efficiency of S. platensis against RA. Serum metabolomics was adopted to profile the potential biomarkers and their corresponding metabolic pathways modulated by Spirulina through UPLC-MS/MS analysis integrated with chemometrics and MetaboAnalyst 5.0 pathway analysis. The results demonstrated that Spirulina exerted significantly modulatory effects in the CFA model by reducing systemic manifestations of oxidative stress, inflammation, and impaired liver and kidney functions typically exemplified by catalase (CAT), superoxide dismutase (SOD), reduced glutathione (GSH), rheumatoid factor (RF), monocyte chemotactic protein 1 (MCP-1), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6), as well as alanine aminotransferase (ALT), aspartate aminotransferase (AST), creatinine, and urea. Histopathological investigations have revealed that Spirulina intervention causes moderately lower inflammatory cells infiltrations, synovial hyperplasia, and cartilage destruction. Regarding serum metabolomics, Spirulina could remarkably reverse disordered RA-associated metabolites, namely glutamic acid, arachidonic acid, 5-hydroxyeicosatetraenoic acid, (20:4/18:0) phosphatidylcholine, and citric acid, to a normal-like state through modulating arachidonic acid metabolism, alanine, aspartate and glutamate metabolism, and citrate cycle pathways putatively implicated in inflammation and joint damage. Our findings provide compelling evidence that S. platensis possesses a broad spectrum of mechanisms to restore the disrupted homeostasis in RA by multi-targeted, synergistic actions.
Collapse
Affiliation(s)
- Dina S Ghallab
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Eman Shawky
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Asmaa A Khalifa
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Samar S Elblehi
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Mohamed M Mohyeldin
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Reham S Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
10
|
Zhang W, Cui N, Su F, Liu M, Li B, Sun Y, Zeng Y, Yang B, Kuang H, Wang Q. Effects of Rehmanniae Radix Praeparata polysaccharides on LPS-induced immune activation in mice based on gut microbiota, metabolomics and transcriptomics. Int J Biol Macromol 2025; 311:143981. [PMID: 40339850 DOI: 10.1016/j.ijbiomac.2025.143981] [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: 02/27/2025] [Revised: 04/27/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
This study explored the immunomodulatory effects of Rehmanniae Radix Praeparata polysaccharides (RP) on LPS-induced immune activation. RP, characterized as a heteropolysaccharide (6.34 kDa and 4.63 kDa) rich in galactose and glucose, was administered to LPS-challenged BALB/c mice at 25 mg/kg and 50 mg/kg doses. Results showed RP significantly reduced pro-inflammatory cytokines (TNF-α, IL-6), lowered oxidative stress (MDA), and boosted antioxidant enzymes (SOD, GSH-Px). It restored splenic structure, mitigated apoptosis, and suppressed the TNF-α/NF-κB/IL-6 pathway. Metabolomics linked RP to sphingolipid metabolism, while gut microbiota analysis revealed increased beneficial bacteria and elevated SCFAs. Transcriptomics confirmed RP's immune regulation via TNF signaling. These findings demonstrate RP's potential in alleviating immune overactivation by modulating inflammation, gut microbiota, and SCFA production, suggesting therapeutic promise for immune-related diseases.
Collapse
Affiliation(s)
- Wensen Zhang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Na Cui
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Fazhi Su
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Meng Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Biao Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Yuanning Zeng
- Guangdong Engineering Technology Research Center for Standardized Processing of Chinese Materia Medica (Guangdong Pharmaceutical University, School of Chinese Materia Medica), Guangdong 510006, China
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Qiuhong Wang
- Guangdong Engineering Technology Research Center for Standardized Processing of Chinese Materia Medica (Guangdong Pharmaceutical University, School of Chinese Materia Medica), Guangdong 510006, China
| |
Collapse
|
11
|
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
|
12
|
Feto NA, Asuzu P, Wan J, Stentz F, Dagogo-Jack S, Mandal N. Do circulating sphingolipid species correlate with age? A study in a normoglycemic biracial population. Biogerontology 2025; 26:106. [PMID: 40323517 PMCID: PMC12052799 DOI: 10.1007/s10522-025-10244-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025]
Abstract
Sphingolipids (SPLs) are essential membrane lipids with significant bioactive roles involved in various cellular processes, and their alterations have been found to be linked to many diseases, including age-related diseases. However, comprehensive studies on the association of plasma sphingolipids with aging in large, diverse cohorts remain limited. The objective of this study was to investigate the relationship between plasma sphingolipid levels and aging in a cohort of 240 normoglycemic, biracial individuals (Black and White), aged 19-65 years. Using a targeted lipidomics approach, we measured 76 sphingolipid species using liquid chromatography-tandem mass spectrometry (LC-MS/MS) in picomole/mL and determined changes in their levels with age and their correlations with aging. We found significant age-related changes in several sphingolipid species, including ceramide C18:1 and several very long-chain sphingomyelins (VLC SMs), such as C28:1 and C30:1, increases with age, showing a positive correlation. On the other hand, glycosphingolipids (monohexosylceramide, MHC; lactosylceramide, LacCer) and sphingosine (So) showed strong negative correlations with aging. A significant correlation was also observed between the ratios of saturate/monosaturated sphingolipid species with aging. In conclusion, our findings provide novel insights into the dynamic changes of circulating sphingolipids with aging. Specific sphingolipid species, such as Ceramide C18:1 and SM, accumulate with age, while others, including MHC, LacCer, and So decrease. These results suggest that the plasma SPL profile may provide valuable information about healthy aging and age-associated disease conditions.
Collapse
Affiliation(s)
- Naser Aliye Feto
- Departments of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Science Center, 930 Madison Ave., Memphis, TN, 38163, USA
| | - Peace Asuzu
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Jim Wan
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Frankie Stentz
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Sam Dagogo-Jack
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
- General Clinical Research Center, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Nawajes Mandal
- Departments of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Science Center, 930 Madison Ave., Memphis, TN, 38163, USA.
- Research, Memphis VA Medical Center, Memphis, TN, 38104, USA.
| |
Collapse
|
13
|
Tsujimura K, Yakabe M, Kano H, Matsumori N. Apoptosis induction by ceramide derivatives and its potential mechanisms through domain formation. Bioorg Med Chem 2025; 126:118222. [PMID: 40327994 DOI: 10.1016/j.bmc.2025.118222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Abstract
Ceramides, which are recognized as pivotal signaling agents in cell differentiation and proliferation, elicit apoptosis in response to anticancer drugs and stress. Although the 1-hydroxy group of ceramide is considered to play an important role in inducing apoptosis, the detailed mechanism underlying ceramide-induced apoptosis remains elusive. We previously synthesized ceramide derivatives by transforming the 1-hydroxy group into amino and carboxy groups, and assessed their ability to form domains in artificial membranes. In this study, we evaluated the apoptotic activities of these derivatives against living cells. Surprisingly, despite the lack of the 1-hydroxy group, most of the derivatives exhibited apoptotic activity, with some being more active than ceramide. Confocal microscopy using fluorescent-ceramide and coherent anti-Stokes Raman scattering microscopy utilizing deuterated ceramide suggested the formation of large domains on living cell membranes. These findings suggest a potential relationship between domain formation and apoptotic activity. Liquid chromatography-mass spectrometry showed that the differences in apoptotic activity among the derivatives were unlikely attributed to variations in cellular uptake. Consequently, we propose that these ceramide derivatives accumulate and form distinct domains within the cellular membranes, inducing apoptosis. Intracellular ceramides synthesized in response to external stimuli may trigger apoptosis through the same mechanism.
Collapse
Affiliation(s)
- Koya Tsujimura
- Department of Chemistry, Graduate School of Science, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Miho Yakabe
- Department of Chemistry, Graduate School of Science, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Hideaki Kano
- Department of Biosciences and Informatics, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama-shi, Kanagawa 223-8522, Japan
| | - Nobuaki Matsumori
- Department of Chemistry, Graduate School of Science, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan.
| |
Collapse
|
14
|
He Y, Xie K, Yang K, Wang N, Zhang L. Unraveling the Interplay Between Metabolism and Neurodevelopment in Health and Disease. CNS Neurosci Ther 2025; 31:e70427. [PMID: 40365712 PMCID: PMC12076066 DOI: 10.1111/cns.70427] [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: 02/24/2025] [Revised: 04/14/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Neurodevelopment is a multifaceted and tightly regulated process essential for the formation, maturation, and functional specialization of the nervous system. It spans critical stages, including cellular proliferation, differentiation, migration, synaptogenesis, and synaptic pruning, which collectively establish the foundation for cognitive, behavioral, and emotional functions. Metabolism serves as a cornerstone for neurodevelopment, providing the energy and substrates necessary for biosynthesis, signaling, and cellular activities. RESULTS Key metabolic pathways, including glycolysis, lipid metabolism, and amino acid metabolism, support processes such as cell proliferation, myelination, and neurotransmitter synthesis. Crucial signaling pathways, such as insulin, mTOR, and AMPK, further regulate neuronal growth, synaptic plasticity, and energy homeostasis. Dysregulation of these metabolic processes is linked to a spectrum of neurodevelopmental disorders, including autism spectrum disorders (ASDs), intellectual disabilities, and epilepsy. CONCLUSIONS This review investigates the intricate interplay between metabolic processes and neurodevelopment, elucidating the molecular mechanisms that govern brain development and the pathogenesis of neurodevelopmental disorders. Additionally, it highlights potential avenues for the development of innovative strategies aimed at enhancing brain health and function.
Collapse
Affiliation(s)
- Yanqing He
- Department of Neurosurgery, and National Clinical Research Center of Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Department of NeurosurgeryXiangya Hospital, Central South University, Jiangxi (National Regional Center for Neurological Diseases)NanchangChina
| | - Kang Xie
- Department of Neurosurgery, and National Clinical Research Center of Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Department of NeurosurgeryXiangya Hospital, Central South University, Jiangxi (National Regional Center for Neurological Diseases)NanchangChina
| | - Kang Yang
- Department of Neurosurgery, and National Clinical Research Center of Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Department of NeurosurgeryXiangya Hospital, Central South University, Jiangxi (National Regional Center for Neurological Diseases)NanchangChina
| | - Nianhua Wang
- Department of NeurosurgeryChangde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City)ChangdeHunanChina
| | - Longbo Zhang
- Department of Neurosurgery, and National Clinical Research Center of Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
- Department of NeurosurgeryXiangya Hospital, Central South University, Jiangxi (National Regional Center for Neurological Diseases)NanchangChina
| |
Collapse
|
15
|
Lin H, Ma C, Cai K, Guo L, Wang X, Lv L, Zhang C, Lin J, Zhang D, Ye C, Wang T, Huang S, Han J, Zhang Z, Gao J, Zhang M, Pu Z, Li F, Guo Y, Zhou X, Qin C, Yi F, Yu X, Kong W, Jiang C, Sun JP. Metabolic signaling of ceramides through the FPR2 receptor inhibits adipocyte thermogenesis. Science 2025; 388:eado4188. [PMID: 40080544 DOI: 10.1126/science.ado4188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 09/13/2024] [Accepted: 01/03/2025] [Indexed: 03/15/2025]
Abstract
Ceramides play a central role in human health and disease, yet their role as systemic signaling molecules remain poorly understood. In this work, we identify formyl peptide receptor 2 (FPR2) as a membrane receptor that specifically binds long-chain ceramides (C14 to C20). In brown and beige adipocytes, C16:0 ceramide binding to FPR2 inhibits thermogenesis through Gi cyclic adenosine monophosphate signaling pathways, an effect that is reversed in the absence of FPR2. We present three cryo-electron microscopy structures of FPR2 in complex with Gi trimers bound to C16:0, C18:0, and C20:0 ceramides. The hydrophobic tails are deeply embedded in the orthosteric ligand pocket, which has a limited amount of plasticity. Modification of the ceramide binding motif in closely related receptors, such as FPR1 or FPR3, converts them from inactive to active ceramide receptors. Our findings provide a structural basis for adipocyte thermogenesis mediated by FPR2.
Collapse
Affiliation(s)
- Hui Lin
- New Cornerstone Science Laboratory, Advanced Medical Research Institute, and NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, Shandong, China
| | - Chuanshun Ma
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Kui Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Lulu Guo
- New Cornerstone Science Laboratory, Advanced Medical Research Institute, and NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Membrane Receptor Drug Target Discovery and Lead Drug Screening at Shandong Province, Shandong, China
| | - Xuemei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Lin Lv
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chao Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jun Lin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Daolai Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Chuan Ye
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Tengwei Wang
- New Cornerstone Science Laboratory, Advanced Medical Research Institute, and NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shenming Huang
- New Cornerstone Science Laboratory, Advanced Medical Research Institute, and NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jifei Han
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Zihao Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Junyan Gao
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration, Jinan, Shandong, China
| | - Mingxiang Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Zhao Pu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
- Department of Biochemistry and Human Biology, University of Toronto, Toronto, Ontario, Canada
| | - Fengyang Li
- School of Pharmacy, Shandong University, Jinan, Shandong, China
| | - Yongyuan Guo
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaojun Zhou
- School of Pharmacy, Shandong University, Jinan, Shandong, China
| | - Chengxue Qin
- School of Pharmacy, Shandong University, Jinan, Shandong, China
| | - Fan Yi
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Jin-Peng Sun
- New Cornerstone Science Laboratory, Advanced Medical Research Institute, and NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Biophysics, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
16
|
Huang FQ, Wang HF, Yang T, Yang D, Liu P, Alolga RN, Ma G, Liu B, Pan A, Liu SJ, Qi LW. Ceramides increase mitochondrial permeabilization to trigger mtDNA-dependent inflammation in astrocytes during brain ischemia. Metabolism 2025; 166:156161. [PMID: 39956315 DOI: 10.1016/j.metabol.2025.156161] [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: 10/08/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
The brain is rich in lipids, and disorders or abnormalities in lipid metabolism can induce neurotoxicity. Ceramides are the central intermediates of sphingolipid metabolism. This study was designed to investigate the potential lipotoxicity of ceramides in brain ischemia. First, a pseudo-targeted lipidomics analysis of plasma samples from stroke patients found significantly elevated levels of long-chain ceramides. A similar observation was made in mice subjected to permanent middle cerebral artery occlusion (pMCAO) surgery. In cultured cells, it was found that the altered ceramides were mainly derived from astrocytes via de novo pathway, and SPTLC2 was a key regulator because Sptlc2 knockdown largely blocked ceramide production. Ceramides induced astrocyte activation and triggered oxidative stress to impair mitochondrial homeostasis by increasing mitochondrial permeabilization. Moreover, ceramides triggered the formation of voltage-dependent anion channel (VDAC) oligomers in the mitochondrial outer membrane, through which mtDNA was released into the cytoplasm. Similar to oxygen and glucose depletion treatment, ceramides also increased cGAS activity and STING protein expression. However, this activity was diminished in the presence of the mitochondrial ROS scavenger SKQ1, indicating the involvement of oxidative stress in ceramide action. By facilitating cGAS/STING signaling cascades, ceramides resultantly induced interferon response to aggravate inflammatory damage in the ischemic brain. To address the impact of ceramides on brain ischemic injury in vivo, ceramide generation was blocked in the brain by injection of AAV9-Sptlc2 shRNA in pMCAO mice. Sptlc2 knockdown in the brain reduced ceramide generation and attenuated brain ischemic damage with astrocyte inactivation. As expected, Sptlc2 deficiency effectively blocked cGAS/STING pathway-dependent interferon responses. Together, these findings suggest a new therapeutic strategy for pharmacological intervention to attenuate neuroinflammation.
Collapse
Affiliation(s)
- Feng-Qing Huang
- Department of Cardiology, Pukou Hospital of Chinese Medicine Affiliated to China Pharmaceutical University, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China; Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| | - Hong-Fei Wang
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, Jiangsu 211198, China; Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Tong Yang
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Dai Yang
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Peian Liu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Raphael N Alolga
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Gaoxiang Ma
- Department of Cardiology, Pukou Hospital of Chinese Medicine Affiliated to China Pharmaceutical University, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Baolin Liu
- Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - An Pan
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| | - Shi-Jia Liu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu 210029, China.
| | - Lian-Wen Qi
- Department of Cardiology, Pukou Hospital of Chinese Medicine Affiliated to China Pharmaceutical University, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China; Clinical Metabolomics Center, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
17
|
Ruan C, Sun J, Liang X, Huang H, Zhang M, Zhang S. Associations of Palmito-leic Acid and Nervonic Acid Hexosylceramides with Type 2 Diabetes Mellitus in a Prospective Nested Case-control Study Among Chinese Population. Endocr Res 2025; 50:109-117. [PMID: 40088080 DOI: 10.1080/07435800.2025.2479256] [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: 11/27/2024] [Revised: 02/05/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVES We aimed to evaluate the associations of hexosylceramides (HexCers) and their ratio with incident type 2 diabetes mellitus (T2DM) and explore underlying mechanisms. METHODS We conducted a nested case-control study using the Suzhou chronic disease cohort including 234 T2DM cases and 468 controls, 1:2 matched on age (±2 y) and sex. HexCer(d18:1/16:1) and HexCer(d18:1/24:1) were measured by targeted UPLC-MS/MS. Multivariable conditional logistic regression was used to estimate the associations of these HexCer species and their ratio with T2DM risk. RESULTS After adjustment for potential confounders, compared with the lowest quartile, the highest quartile of HexCer(d18:1/24:1) was positively associated with T2DM risk (OR: 1.91; 95%CI, 1.12, 3.26; P-trend <0.05). The ratio of HexCer(d18:1/24:1) to HexCer(d18:1/16:1) showed a positive association with T2DM risk (OR: 1.89; 95%CI, 1.13, 3.18; P-trend <0.05). On the natural log scale, each SD increases in HexCer(d18:1/24:1) and its ratio to HexCer(d18:1/16:1) increased by 29% and 30%, respectively. No significant association for HexCer(d18:1/16:1) was found. Additive value of HexCer(d18:1/24:1) or HexCer(d18:1/24:1)/HexCer(d18:1/16:1) ratio for prediction of T2DM above traditional risk factors. CONCLUSIONS HexCer(d18:1/24:1) and its ratio to HexCer(d18:1/16:1) are positively associated with incident T2DM in a community-based Chinese population. Future studies are warranted to confirm our findings.
Collapse
Affiliation(s)
- Chuyi Ruan
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Jun Sun
- Department of Medical Service, Kunshan First People's Hospital, Kunshan, Jiangsu, China
| | - Xiaoqing Liang
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Hong Huang
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Mingzhi Zhang
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| | - Shaoyan Zhang
- Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
18
|
Jin S, Zhang Y, Fu H, Zhang W, Qiao H, Xiong Y, Jiang S. Transcriptome Profiling Analysis Reveals Changes in the Antioxidant Defense System, Morphology, and Gene Expression in the Gills of Macrobrachium nipponense Caused by Alkalinity Exposure. Int J Mol Sci 2025; 26:4321. [PMID: 40362560 PMCID: PMC12072836 DOI: 10.3390/ijms26094321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
The median lethal concentration value of alkalinity tolerance for Macrobrachium nipponense over 96 h is only 14.42 mmol/L with a safety value of 4.71 mmol/L, which is insufficient to perform the aquaculture program in a water environment with high alkalinity. Thus, the present study aims to explore the effects of alkalinity exposure on the gills of M. nipponense through identifying the changes in antioxidant enzymes, morphology, and gene expressions after 1 day, 4 days, and 7 days of exposure under an alkalinity of 10 mmol/L. The activities of MDA, GSH-PX, CAT, T-AOC, and Ca2+Mg2+-ATPase are significantly stimulated by 62.6%, 6.57%, 32.1%, 33.3%, and 14.9%, compared to those from Day 0 (control group), indicating that these antioxidant enzymes play essential roles in the protection of prawns from the damage of reactive oxygen species caused by alkalinity exposure. In addition, alkalinity exposure results in an increase in the hemolymph vessels, affecting the normal respiratory function of the gills. Transcriptome profiling analysis reveals that short-term alkali exposure (4 days) does not result in significant changes in gene expression in the present study. Furthermore, metabolic pathways, biosynthesis of amino acids, amino sugar and nucleotide sugar metabolism, lysosomes, glycolysis/gluconeogenesis, and phagosomes represent the main enriched metabolic pathways of differentially expressed genes (DEGs) between Day 4 and Day 7. Biosynthesis of amino acids, lysosomes, and phagosomes are immune-related metabolic pathways, while amino sugar and nucleotide sugar metabolism and glycolysis/gluconeogenesis are energy metabolism-related metabolic pathways, indicating that the processes of immune response and energy metabolism play essential roles in the response to alkalinity exposure in M. nipponense. Thus, the DEGs from these metabolic pathways are considered as candidate genes involved in the regulation of alkaline acclimation in M. nipponense. The present study provides valuable evidence for analysis of the adaptive mechanism when exposed to alkalinity, contributing to the survival rate and aquaculture of this species under water environments with high alkalinity.
Collapse
Affiliation(s)
- Shubo Jin
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China (W.Z.)
- Key Laboratory of Mariculture & Stock Enhancement in North China’s Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Yuefan Zhang
- Key Laboratory of Mariculture & Stock Enhancement in North China’s Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Hongtuo Fu
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China (W.Z.)
| | - Wenyi Zhang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China (W.Z.)
| | - Hui Qiao
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China (W.Z.)
- Key Laboratory of Mariculture & Stock Enhancement in North China’s Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| | - Yiwei Xiong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China (W.Z.)
| | - Sufei Jiang
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China (W.Z.)
- Key Laboratory of Mariculture & Stock Enhancement in North China’s Sea, Ministry of Agriculture and Rural Affairs, Dalian Ocean University, Dalian 116023, China
| |
Collapse
|
19
|
Niu Z, Ako AA, Geiger SD, Howe CG, Perng W, Singh R, Karagas MR, Elliott AJ, Cassidy-Bushrow A, Camargo CA, Sanderson K, McEvoy CT, Oken E, Dabelea D, Hartert TV, Carter B, Stroustrup A, Lampland A, O’Connor TG, Gogcu S, Hudak ML, Shorey-Kendrick LE, Zhao Q, Ni Y, VanWormer J, Ferrara A, Hedderson M, Zhu Y, Alshawabkeh A, Cordero J, Koinis-Mitchell D, Carnell S, Breton CV, Bastain TM, Farzan SF. Maternal Cardiometabolic Risk Factors in Pregnancy and Offspring Blood Pressure at Age 2 to 18 Years. JAMA Netw Open 2025; 8:e259205. [PMID: 40338548 PMCID: PMC12062903 DOI: 10.1001/jamanetworkopen.2025.9205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/10/2025] [Indexed: 05/09/2025] Open
Abstract
Importance Higher blood pressure in early life may signal cardiovascular disease over the life course, but determinants of blood pressure in early life are poorly understood. Objective To examine the association of maternal cardiometabolic risk factors during pregnancy with offspring blood pressure from age 2 to 18 years and explore whether the association is modified by offspring sex and race and ethnicity. Design, Setting, and Participants This cohort study analyzed data from the Environmental Influences on Child Health Outcomes program between January 1, 1994, and March 31, 2023. Three common maternal cardiometabolic risk factors during pregnancy were examined: prepregnancy obesity, gestational diabetes, and hypertensive disorders of pregnancy (HDP). Exposure Maternal cardiometabolic risk factors were retrieved and harmonized from medical records and questionnaires. Main Outcomes and Measures Offspring systolic blood pressure (SBP) and diastolic blood pressure (DBP) percentiles adjusted for age, sex, and height were calculated. Results Among 12 480 mother-offspring pairs (mean [SD] maternal age during pregnancy, 29.9 [6.4] years; 856 of 12 303 identifying as Asian [7.0%]; 1908 as Black [15.5%]; 2305 as Hispanic [18.7%]; 6522 as White [52.3%], and 712 as other [5.8%] race and ethnicity), at least 1 maternal cardiometabolic risk factor was present in 5537 (44.4%), with prepregnancy obesity being the most prevalent (3072 [24.6%]), followed by HDP (1693 [13.6%]) and gestational diabetes (805 [6.5%]). Offspring born to mothers with any cardiometabolic risk factors had higher SBP (4.88 percentile points; 95% CI, 3.97-5.82 percentile points) and higher DBP (1.90 percentile points; 95% CI, 1.15-2.64 percentile points) at their first blood pressure measurement, after adjusting for potential confounders, compared with their counterparts without any risk factors. Hypertensive disorders of pregnancy, alone or with either prepregnancy obesity or gestational diabetes, was significantly associated with higher offspring blood pressure. These associations were generally more significant among female compared with male offspring and among Black compared with other racial and ethnic groups. Among 6015 offspring who had 2 or more blood pressure measures, maternal cardiometabolic risk factors were associated with an increased rate of blood pressure change from age 2 to 18 years (SBP percentile, 0.5 [95% CI, 0.2-0.8] per year; DBP percentile, 0.7 [95% CI 0.5-1.0] per year). Conclusions and Relevance These findings suggest that protecting pregnant individuals from cardiometabolic risk factors may promote healthier blood pressure in the next generation.
Collapse
Affiliation(s)
- Zhongzheng Niu
- Division of Environmental Health, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, State University of New York at Buffalo
| | - Ako Adams Ako
- Department of Pediatrics, Children’s Hospital at Montefiore, Bronx, New York
| | - Sarah Dee Geiger
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign
| | - Caitlin G. Howe
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire
| | - Wei Perng
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora
| | - Rachana Singh
- Department of Pediatrics, Tufts University School of Medicine, Boston, Massachusetts
| | - Margaret R. Karagas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, New Hampshire
| | | | - Andrea Cassidy-Bushrow
- Department of Public Health Sciences, Henry Ford Health, Detroit, Michigan
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing
| | - Carlos A. Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Keia Sanderson
- Department of Medicine, University of North Carolina at Chapel Hill
| | - Cynthia T. McEvoy
- Department of Pediatrics, Papé Pediatric Research Institute, Oregon Health & Science University, Portland
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts
| | - Dana Dabelea
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora
| | - Tina V. Hartert
- Departments of Medicine and Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Brian Carter
- Department of Pediatrics-Neonatology, Children’s Mercy Hospital, Kansas City, Missouri
| | - Annemarie Stroustrup
- Division of Neonatology, Department of Pediatrics, Cohen Children’s Medical Center at Northwell Health, New York, New York
| | | | - Thomas G. O’Connor
- Department of Psychiatry, University of Rochester Medical Center, Rochester, New York
| | - Semsa Gogcu
- Division of Neonatology, Department of Pediatrics, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Mark L. Hudak
- Department of Pediatrics, University of Florida College of Medicine, Gainesville
| | - Lyndsey E. Shorey-Kendrick
- Department of Pediatrics, Papé Pediatric Research Institute, Oregon Health & Science University, Portland
| | - Qi Zhao
- Department of Preventative Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis
| | - Yu Ni
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle
- School of Public Health, San Diego State University, San Diego, California
| | | | - Assiamira Ferrara
- Division of Research, Kaiser Permanente Northern California, Pleasanton, California
| | - Monique Hedderson
- Division of Research, Kaiser Permanente Northern California, Pleasanton, California
| | - Yeyi Zhu
- Division of Research, Kaiser Permanente Northern California, Pleasanton, California
| | - Akram Alshawabkeh
- Department of Civil and Environmental Engineering, Northeastern University, Boston, Massachusetts
| | - Jose Cordero
- Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens
| | - Daphne Koinis-Mitchell
- Departments of Pediatrics and Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Susan Carnell
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Carrie V. Breton
- Division of Environmental Health, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles
| | - Theresa M. Bastain
- Division of Environmental Health, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles
| | - Shohreh F. Farzan
- Division of Environmental Health, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles
| |
Collapse
|
20
|
Jean Pierre AR, Kasirajan A, Green SR, Sivaprakasam M, Sahaya Raj RS, Josyula JVN, Mutheneni SR, Subramanyam V, Pillai AB. Clinical correlations of plasma sphingosine-1-phosphate and sphingolipid key enzymes in severe dengue using laboratory and machine learning approach. Clin Chim Acta 2025; 574:120335. [PMID: 40306535 DOI: 10.1016/j.cca.2025.120335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 04/05/2025] [Accepted: 04/26/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Sphingolipids are crucial for vascular integrity and cellular homeostasis, with recent studies highlighting their role in viral diseases. OBJECTIVES The study aimed to assess the plasma levels of sphingolipids, specifically Sphingosine-1-phosphate (S1P) and the key enzymes of sphingolipid metabolism: Sphingomyelin synthase (SMS1), Ceramide Kinase (CERK) and acid ceramidase (ASAH1) and its association with clinical outcomes of dengue. METHODS This prospective cohort study had 102 dengue cases with 17 severe dengue (SD), 33 dengue with warning signs (DWW), 52 dengue without warning signs (DWOW) along with 10 each from other febrile illnesses and healthy controls. Blood was collected across febrile, defervescence, and convalescence phases. Plasma levels of S1P and the enzymes were measured using ELISA, mRNA using qRT-PCR. Predictive efficacy was determined using Support vector machine (SVM) models. RESULTS Study showed a significant reduction in S1P levels across all dengue forms during febrile phases, with further decline in SD during the critical phase (P < 0.05).mRNA levels of the enzymes were increasing during critical phase (P ≤ 0.001) with no significant difference noted in their respective protein levels. S1P and SMS1 levels correlated significantly with clinical severity indicators, including hematocrit, albumin, platelet count, and liver enzymes. SVM analysis identified CERK levels along with platelet count, HCT, and ALT as markers with high predictive accuracy for dengue severity. CONCLUSION The study reports an association of sphingolipids with dengue virulence, emphasizing the role of S1P metabolism in disease progression and plasma leakage, and highlighting the potential of targeting sphingolipids in managing severe dengue.
Collapse
Affiliation(s)
- Aashika Raagavi Jean Pierre
- MGM Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607402, India
| | - Anand Kasirajan
- MGM Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607402, India
| | - Siva Ranganathan Green
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India
| | - Manikandan Sivaprakasam
- MGM Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607402, India
| | - Rithanya Syam Sahaya Raj
- MGM Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607402, India
| | | | - Srinivasa Rao Mutheneni
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
| | - Veni Subramanyam
- MGM Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607402, India
| | | |
Collapse
|
21
|
Kalkman HO, Smigielski L. Ceramides may Play a Central Role in the Pathogenesis of Alzheimer's Disease: a Review of Evidence and Horizons for Discovery. Mol Neurobiol 2025:10.1007/s12035-025-04989-0. [PMID: 40295359 DOI: 10.1007/s12035-025-04989-0] [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] [Accepted: 04/19/2025] [Indexed: 04/30/2025]
Abstract
While several hypotheses have been proposed to explain the underlying mechanisms of Alzheimer's disease, none have been entirely satisfactory. Both genetic and non-genetic risk factors, such as infections, metabolic disorders and psychological stress, contribute to this debilitating disease. Multiple lines of evidence indicate that ceramides may be central to the pathogenesis of Alzheimer's disease. Tumor necrosis factor-α, saturated fatty acids and cortisol elevate the brain levels of ceramides, while genetic risk factors, such as mutations in APP, presenilin, TREM2 and APOE ε4, also elevate ceramide synthesis. Importantly, ceramides displace sphingomyelin and cholesterol from lipid raft-like membrane patches that connect the endoplasmic reticulum and mitochondria, disturbing mitochondrial oxidative phosphorylation and energy production. As a consequence, the flattening of lipid rafts alters the function of γ-secretase, leading to increased production of Aβ42. Moreover, ceramides inhibit the insulin-signaling cascade via at least three mechanisms, resulting in the activation of glycogen synthase kinase-3 β. Activation of this kinase has multiple consequences, as it further deteriorates insulin resistance, promotes the transcription of BACE1, causes hyperphosphorylation of tau and inhibits the transcription factor Nrf2. Functional Nrf2 prevents apoptosis, mediates anti-inflammatory activity and improves blood-brain barrier function. Thus, various seemingly unrelated Alzheimer's disease risk factors converge on ceramide production, whereas the elevated levels of ceramides give rise to the well-known pathological features of Alzheimer's disease. Understanding and targeting these mechanisms may provide a promising foundation for the development of novel preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Hans O Kalkman
- Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Lukasz Smigielski
- Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Yang B, Chen S, Xia X, Tao Z, Liu C, Li S, Zhang S, Huang J, Xia L, Quan W, Yang C, Li J. Mas Signaling Potentiates Neutropil Extracellular Traps Formation Induced by Endothelial Cells Derived S1P in Mice with Acute Liver Failure. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2411428. [PMID: 40285622 DOI: 10.1002/advs.202411428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 03/31/2025] [Indexed: 04/29/2025]
Abstract
Mas, a newly identified G-protein-coupled receptor, is prevalent in myeloid-derived immune cells and plays a key role in inflammation. This study investigates Mas signaling and neutrophil extracellular traps (NETs) in acute liver failure (ALF), aiming to elucidate their mechanisms. Male Mas1-/- and wild-type mice, aged 6-8 weeks, receive intraperitoneally injected with lipopolysaccharide (LPS)/D-galactosamine (D-Gal) (L/G) to study NETs formation. Hepatic Mas expression increases in WT-L/G mice, whereas systemic Mas1 knockout significantly reduces L/G-induced NETs and hepatotoxicity. Antibiotics treatment and co-housing (Mas1-/--L/G and WT-L/G mice) experiments show that gut flora influences the disease phenotype in Mas1-/--L/G mice. Fecal metabolite analysis suggests that mice may be protected by reduced deoxycholic acid (DCA) production in Mas1-/- activated hepatic farnesoid X receptor (FXR), suppressing sphingosine-1-phosphate (S1P)-dependent NETs. Additionally, Mas1-/- also activates the FXR-S1P-NETs axis in the liver by inhibiting SHP2. Single-cell sequencing shows decreased interaction between endothelial cells and Cldn1+CD177+ senescent neutrophils through Col4a1-CD44. This inhibits S1P-induced Raf signaling pathway activation and NETs formation. Mas signaling significantly impacts NETs formation, highlighting its potential as an anti-inflammatory therapeutic target for ALF.
Collapse
Affiliation(s)
- Bo Yang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shuai Chen
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Xiaoqi Xia
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Ziwen Tao
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Chun Liu
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shanshan Li
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Shuo Zhang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jiali Huang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Lu Xia
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Wenqiang Quan
- Department of Laboratory Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Changqing Yang
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jing Li
- Department of Gastroenterology and Hepatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| |
Collapse
|
23
|
Kondraciuk M, Chlabicz M, Jamiołkowski J, Zieleniewska N, Ciborowski M, Godlewski A, Sawicka-Śmiarowska E, Ptaszyńska K, Łapińska M, Krętowski A, Kamiński KA. Coronary artery disease is associated with particular change of serum metabolome: a case-control study. Metabolomics 2025; 21:57. [PMID: 40281287 PMCID: PMC12031763 DOI: 10.1007/s11306-025-02253-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 04/02/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Cardiovascular disease (CVD) is a significant cause of mortality worldwide. Preventive programs are trying to reduce the burden of the disease. Recent advances in metabolomics profiling open a new avenue for developing complementary CVD evaluation strategies. OBJECTIVES The aim of the study was to investigate whether a metabolomic profile can provide an additional characterisation of individuals with coronary artery disease (CAD). METHODS The study included 167 participants with CAD aged 41-79 years. A control group was formed of 166 individuals without CAD, gender- and age-matched to the study group. A total of 188 metabolites were profiled in serum by liquid chromatography-tandem mass spectrometry. After clearing the data, associations between 132 metabolites and CAD presence were analysed using multiple linear regression models. RESULTS We observed significant differences in serum metabolic profiles between analysed groups on various levels. However, a deeper analysis revealed sphingomyelin 41:1 (SM 41:1) as the main metabolite independently associated with CAD after correction for classical CV risk factors. Its concentration was lower in the CAD group (median 9.79 µmol/L, interquartile range (IQR) 7.92-12.23) compared to control one (median 13.60 µmol/L, IQR 11.30-16.15) (p < 0.001). Further analysis showed that SM 41:1 concentration was inversely correlated with CAD, current smoking, and hypertension; and positively associated with female gender and non-HDL level. CONCLUSIONS CAD patients present lower plasma concentrations of SM 41:1 than healthy subjects. A better understanding of the biological function of sphingomyelin in CAD patients may help develop therapeutic approaches and risk stratification in this group.
Collapse
Affiliation(s)
- Marcin Kondraciuk
- Population Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Małgorzata Chlabicz
- Population Research Centre, Medical University of Bialystok, Bialystok, Poland.
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland.
- Department of Invasive Cardiology, Medical University of Bialystok, Bialystok, Poland.
| | - Jacek Jamiołkowski
- Population Research Centre, Medical University of Bialystok, Bialystok, Poland
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Natalia Zieleniewska
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland
| | - Michał Ciborowski
- Metabolomics and Proteomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Godlewski
- Metabolomics and Proteomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | | | | | - Magda Łapińska
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Adam Krętowski
- Metabolomics and Proteomics Laboratory, Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Karol A Kamiński
- Population Research Centre, Medical University of Bialystok, Bialystok, Poland
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
- Department of Cardiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
24
|
Shi W, Lin H, Di W, He C, Shen Y. Granulosa cell RNA-Seq insights into senescence and sphingolipid metabolism disorder in PCOS: aspirin as a potential therapeutic drug. Reprod Biol Endocrinol 2025; 23:61. [PMID: 40287692 PMCID: PMC12032776 DOI: 10.1186/s12958-025-01396-x] [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: 12/30/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a pivotal cause of anovulatory infertility and the pathogenesis remains elusive. Cellular senescence and sphingolipid metabolism disorder are closely intertwined, and both have been demonstrated present within the granulosa cells of PCOS, while research on the combined impact of senescence and sphingolipids on PCOS-related anovulation is scarce. METHODS Here, we leveraged four datasets of PCOS and executed differential gene expression analysis, engaged in WGCNA, and harnessed machine learning algorithms-including RF, SVM-RFE, and LASSO-to deeply explore the key genes that interact with senescence and sphingolipid metabolism in granulosa cells of PCOS. These key genes were subjected to further analysis to construct a diagnostic model, forecast immune cell infiltration, and identify potential agents. Additionally, within the testosterone-stimulated granulosa cells, we validated the expression of key genes, confirmed senescence and sphingolipids dysregulation, and evaluated the therapeutic efficacy of the candidate agent. RESULTS Our research pinpointed a set of genes (LYN, PLCG2, STAT5B, MMP9, and IL6R) that showed promise as biomarkers for PCOS-related anovulation and the diagnostic nomogram was developed. These biomarkers were linked to various immune cell types infiltration. In testosterone-stimulated granulosa cells, we observed increased expression of these biomarkers, accompanied by signs of senescence and changes in sphingolipids. Importantly, the potential agent aspirin displayed the ability to ameliorate these two processes. CONCLUSION This study highlighted the important value of genes associated with senescence and sphingolipids dysregulation in PCOS. Aspirin targeting senescence could be a promising therapeutic drug for addressing anovulation associated with PCOS.
Collapse
Affiliation(s)
- Weiwei Shi
- Department of Obstetrics and Gynecology, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, Jiangsu, China
| | - Hao Lin
- Department of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
- Department of Clinical Science and Research, Zhongda Hospital Affiliated to Southeast University, Nanjing, 21009, Jiangsu, China
| | - Wu Di
- Department of Obstetrics and Gynecology, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, Jiangsu, China
| | - Cong He
- Key Laboratory of Innovative Applications of Bioresources and Functional Molecules of Jiangsu Province, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing, 210013, Jiangsu, China
| | - Yang Shen
- Department of Obstetrics and Gynecology, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, Jiangsu, China.
- Department of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
25
|
Meng W, Yan J, Zhao Y, Ye Z, Ma X, Wei W, Chen J, Zhang L. Identifying the specific lipid biomarkers and LYPLA1 as a novel candidate in intramuscular fat deposition of Erhualian pigs. BMC Genomics 2025; 26:407. [PMID: 40281449 PMCID: PMC12032779 DOI: 10.1186/s12864-025-11611-z] [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: 10/18/2024] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Intramuscular fat (IMF) is a key determinant of meat quality enhancement in pigs. However, its correlation with subcutaneous fat (SCF) deposition presents a challenge. The precise regulation of IMF lipogenesis, without impacting SCF deposition, is a critical issue in the pig industry. To investigate this, our study examined the lipid profiles of longissimus dorsi (LD) muscle and subcutaneous adipose tissue in high IMF (IMFH) and low IMF (IMFL) Chinese Erhualian pigs using lipidomics techniques. RESULTS We identified 112 differentially abundant lipids (DALs) in the muscle and 101 DALs in the adipose tissue. Notably, 105 specific DALs associated with IMF, including various candidate markers like upregulated lipids of PS (19:2/18:1), TG (14:2/4:0/4:0), PS (17:1/18:2), FA(10:0) + COOH:(s), CL (20:4/18:2/18:2/18:2), and downregulated lipids of FA (20:4), SM (d43:5), TG (38:1/22:6), PC (22:3/20:3), and PC (18:2/24:8), were identified. These specific DALs were implicated in the regulation of linoleic, arachidonic, and alpha-linolenic acid metabolism, and choline metabolism in cancer. We further discovered that the lysophosphlipase 1 (LYPLA1) gene promotes differentiation and lipid deposition of intramuscular pre-adipocytes by affecting the phosphatidylcholine (PC) content. CONCLUSIONS Our findings identify specific lipids associated with IMF accumulation in both skeletal muscle and subcutaneous adipose depots, thereby offering valuable insights into the lipid composition of porcine IMF and new avenues for targeted IMF deposition.
Collapse
Affiliation(s)
- Wenjing Meng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - JiaYu Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yuelei Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zijian Ye
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiangfei Ma
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jie Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Lifan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Lee EJ, Park S, Jeong KS. Sirt2 deficiency aggravates intramuscular adipose tissue infiltration and impairs myogenesis with aging in male mice. Biogerontology 2025; 26:93. [PMID: 40257511 DOI: 10.1007/s10522-025-10238-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/08/2025] [Indexed: 04/22/2025]
Abstract
Sarcopenia, closely associated with other diseases such as diabetes, metabolic syndrome, and osteoporosis, significantly impacts aging populations. It is characterized by muscle atrophy, increased intramuscular adipose tissue, impaired myogenesis, chronic low-grade inflammation, and reduced muscle function. The mechanisms behind aging muscle remain incompletely understood. This study aims to elucidate the role of Sirt2 in the aging process of skeletal muscles and enhance our understanding of the underlying mechanisms. Sirt2 expression was reduced in aging muscle of male mice by 40%, compared to young muscle. Aged male Sirt2 knockout mice exhibit increased intramuscular adipose tissue infiltration by 8.5-fold changes. Furthermore, the deletion of Sirt2 exacerbated myogenesis impairment in aged muscle by decreasing the expression of Pax7 (50%) and NogoA (80%), compared to age- and sex- matched counterparts, emphasizing the role of Sirt2 in pathology of aging muscle. Additionally, long-term Sirt2 deletion affected other Sirtuin subfamily members, with decreased expressions of Sirt1 (65%), Sirt4 (94%), and Sirt5 (71%), and increased expressions of Sirt6 (4.6-fold) and Sirt7 (2.8-fold) in old male Sirt2 knockout mice, while there was no difference of these gene expression in young male mice. This study underscores the critical need for a deeper investigation into Sirt2, promising new insights that could lead to targeted therapies for sarcopenia, ultimately improving the quality of life in the elderly.
Collapse
Affiliation(s)
- Eun-Joo Lee
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, 02115, USA.
| | - SunYoung Park
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Kyu-Shik Jeong
- Department of Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Department of Companion Animal Health, Daegu Haany University, Gyeongsan, 38610, Republic of Korea.
- Stellamed Co., LTD, Daegu, 41504, Republic of Korea.
| |
Collapse
|
29
|
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
|
30
|
Erskine D, Bronowska AK, Outeiro TF, Attems J. Sphingolipidoses: expanding the spectrum of α-synucleinopathies. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02925-z. [PMID: 40244388 DOI: 10.1007/s00702-025-02925-z] [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: 03/06/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
Although α-synuclein pathology is typically associated with Lewy body diseases and multiple systems atrophy, increasing evidence indicates that it also occurs in a group of lysosomal storage disorders termed sphingolipidoses caused by the incomplete degradation, and subsequent accumulation, of a class of lipids termed sphingolipids. Notably, a number of genes that cause sphingolipidoses are also risk genes for Lewy body diseases, suggesting aetiological links between these distinct disorders. In the present review, we discuss the sphingolipidoses in which α-synuclein pathology has been reported: Gaucher disease, Krabbe disease, metachromatic leukodystrophy, Tay-Sachs disease and Anderson-Fabry disease, and describe the characteristic clinical and pathological features of these disorders, in addition to the evidence suggesting α-synuclein pathology occurs in these disorders. Finally, we evaluate the pathological mechanisms that underlie these rare disorders, with particular attention to how the enzymatic deficiency, substrate accumulation, or both, could contribute to the genesis of α-synuclein pathology and the implications of this for Lewy body diseases.
Collapse
Affiliation(s)
- Daniel Erskine
- Metabolic Neurodegeneration Laboratory, Newcastle University, Newcastle, UK.
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK.
| | - Agnieszka K Bronowska
- Chemistry - School of Natural and Environmental Sciences, Newcastle University, Newcastle, UK
| | - Tiago F Outeiro
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
- DZNE, Gottingen, Germany
- University Medical Center Gottingen, Newcastle, Germany
| | - Johannes Attems
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| |
Collapse
|
31
|
Zhakupova A, Zeinolla A, Kokabi K, Sergazy S, Aljofan M. Drug Resistance: The Role of Sphingolipid Metabolism. Int J Mol Sci 2025; 26:3716. [PMID: 40332322 PMCID: PMC12027666 DOI: 10.3390/ijms26083716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/10/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
A significant challenge in cancer treatment is the rising problem of drug resistance that reduces the effectiveness of therapeutic strategies. Current knowledge shows that multiple mechanisms play a role in cancer drug resistance. Another mechanism that has gained attention is the alteration in sphingolipid trafficking and the dysregulation of its metabolism, which was reported to cause cancer-associated drug resistance. Sphingolipids are lipids containing sphingosine and have multiple roles, ranging from lipid raft formation, apoptosis, and cell signaling to immune cell trafficking. Recent studies show that in developing cancer cells, altered or dysregulated sphingolipids are associated with drug efflux and promote the survival of cancer cells by bypassing apoptosis. Upregulated levels of the glucosylceramide synthase (GCS), an enzyme that functions in sphingolipid metabolism, lead to the upregulated ABCB1 gene that induces drug efflux from the cancer cells. These bypass mechanisms make drugs that induce apoptosis in tumor cells ineffective. By highlighting the current findings, this review aims to provide a mechanism of drug resistance caused by the dysregulation of glucosylceramide synthase, sphingosine kinase, and acid ceramidase enzymes as possible therapeutic targets to enhance the effectiveness of the currently used chemotherapeutic agents.
Collapse
Affiliation(s)
- Assem Zhakupova
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan; (A.Z.); (A.Z.); (K.K.)
| | - Adelina Zeinolla
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan; (A.Z.); (A.Z.); (K.K.)
| | - Kamilya Kokabi
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan; (A.Z.); (A.Z.); (K.K.)
| | - Shynggys Sergazy
- Drug Discovery and Development Laboratory, National Laboratory Astana, Astana 010000, Kazakhstan
- LLP “VICTUS PHARM”, Astana 010000, Kazakhstan
| | - Mohamad Aljofan
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan; (A.Z.); (A.Z.); (K.K.)
- Drug Discovery and Development Laboratory, National Laboratory Astana, Astana 010000, Kazakhstan
| |
Collapse
|
32
|
Wang R, Gan C, Gong B, Huang J, Lou Z, Wang D, Yan R, Li G, Xiong T, Guo J. Tongfu Xingshen capsule alleviates stroke-associated pneumonia-induced multiple organ injuries by modulating the gut microbiota and sphingolipid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156756. [PMID: 40252432 DOI: 10.1016/j.phymed.2025.156756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/14/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Stroke-associated pneumonia (SAP) represents a major complication and cause of death in patients suffering from intracerebral haemorrhage (ICH). It's urgent to develop more effective therapeutic strategies. Tongfu Xingshen capsule (TFXS) is a traditional Chinese medicine that has been utilised in clinical studies for the treatment of ICH and SAP, but the underlying mechanisms remain to be fully elucidated. PURPOSE This study aims to explore the therapeutic effects and mechanisms of TFXS on SAP using an aspiration-induced Klebsiella pneumoniae infection-complicating ICH rat model and an intratracheal injection of lipopolysaccharide (LPS)-induced acute lung injury-complicating ICH rat model. METHODS The chemical components of TFXS are characterised using ULPLC-Q Exactive-Orbitrap-MS. The therapeutic effects of TFXS are evaluated through neurological scoring, histopathology analysis, magnetic resonance imaging, immunofluorescence, Alcian blue-nuclear fast red staining, myeloperoxidase activity assessment, leukocyte counting, and ELISA. To investigate the underlying mechanisms, faecal microbiota transplantation, 16S rRNA sequencing, untargeted metabolomics, and Spearman correlation analyses are performed. RESULTS A total of 60 compounds are identified in TFXS. Pharmacological analysis reveals that TFXS significantly mitigates neurological deficits, enhances haematoma absorption, attenuates brain damage and neuroinflammation, and improves pneumonia and pulmonary injury by reducing the infiltration of leukocytes and lymphocytes, as well as suppressing the infiltration and overactivation of neutrophils. TFXS also alleviates intestinal lesions and barrier damage by increasing acidic mucins and the expression of the tight junction protein zonula occludens-1 (ZO-1). Mechanistically, TFXS ameliorates pneumonia and pulmonary injury in a gut microbiota-dependent manner. It reverses sphingolipid metabolism disorders and ceramide accumulation by modulating SAP-induced gut microbiota dysbiosis and enhancing the abundance of probiotics, including Lactobacillus, Allobaculum and Enterococcus. CONCLUSION TFXS exerts anti-inflammatory and protective effects on the brain, lung, and gut by alleviating gut microbiota dysbiosis and sphingolipid metabolism disorders. These findings highlight TFXS as a promising therapeutic candidate for the treatment of SAP.
Collapse
Affiliation(s)
- Ruihua Wang
- Research Team of Prevention and Treatment of Cerebral Hemorrhage Applying Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China
| | - Changlian Gan
- School of Traditional Dai Medicine, West Yunnan University of Applied Science, JH, Xishuangbanna, Yunnan Province, 666100, PR China
| | - Baoying Gong
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China
| | - Juan Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China
| | - Zhenzhen Lou
- Research Team of Prevention and Treatment of Cerebral Hemorrhage Applying Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China
| | - Daxiu Wang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, PR China
| | - Geng Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510000, PR China.
| | - Tianqin Xiong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 511400, PR China.
| | - Jianwen Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Department of Neurology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510120, PR China.
| |
Collapse
|
33
|
Rong J, Liang SH. Novel Pyrazolo[1,5- a]pyrimidine-3,5-diamines as nSMase2 Inhibitors. ACS Med Chem Lett 2025; 16:534-535. [PMID: 40236526 PMCID: PMC11995204 DOI: 10.1021/acsmedchemlett.5c00134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
This patent highlights novel pyrazolo[1,5-a]pyrimidine-3,5-diamines as neutral sphingomyelinase 2 (nSMase2) inhibitors. It provides detailed information on pyrazolo[1,5-a]pyrimidine-3,5-diamine compounds, their pharmaceutical composition, use, and potential therapeutic applications for nSMase2-related diseases.
Collapse
Affiliation(s)
- Jian Rong
- Department of Radiology and Imaging
Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Steven H. Liang
- Department of Radiology and Imaging
Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| |
Collapse
|
34
|
Brunmaier LAE, Ozdemir T, Walker TW. Angiogenesis: Biological Mechanisms and In Vitro Models. Ann Biomed Eng 2025:10.1007/s10439-025-03721-2. [PMID: 40210793 DOI: 10.1007/s10439-025-03721-2] [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: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
The translation of biomedical devices and drug research is an expensive and long process with a low probability of receiving FDA approval. Developing physiologically relevant in vitro models with human cells offers a solution to not only improving the odds of FDA approval but also to expand our ability to study complex in vivo systems in a simpler fashion. Animal models remain the standard for pre-clinical testing; however, the data from animal models is an unreliable extrapolation when anticipating a human response in clinical trials, thus contributing to the low rates of translation. In this review, we focus on in vitro vascular or angiogenic models because of the incremental role that the vascular system plays in the translation of biomedical research. The first section of this review discusses the most common angiogenic cytokines that are used in vitro to initiate angiogenesis, followed by angiogenic inhibitors where both initiators and inhibitors work to maintain vascular homeostasis. Next, we evaluate previously published in vitro models, where we evaluate capturing the physical environment for biomimetic in vitro modeling. These topics provide a foundation of parameters that must be considered to improve and achieve vascular biomimicry. Finally, we summarize these topics to suggest a path forward with the goal of engineering human in vitro models that emulate the in vivo environment and provide a platform for biomedical device and drug screening that produces data to support clinical translation.
Collapse
Affiliation(s)
- Laura A E Brunmaier
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Travis W Walker
- Karen M. Swindler Department of Chemical and Biological Engineering, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA.
| |
Collapse
|
35
|
Mohammed S, Kalogeropoulos AP, Alvarado V, Weisfelner-Bloom M, Clarke CJ. Serum and plasma sphingolipids as biomarkers of chemotherapy-induced cardiotoxicity in female patients with breast cancer. J Lipid Res 2025; 66:100798. [PMID: 40189207 DOI: 10.1016/j.jlr.2025.100798] [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: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025] Open
Abstract
Although effective as a chemotherapeutic, the utility of Doxorubicin (Dox) is hampered by cardiotoxicity. Despite this, the ability to predict and guide monitoring of patients receiving Dox is hampered by a lack of effective biomarkers to identify susceptible patients and detect early signs of subclinical cardiotoxicity. Based on their well-established roles in the response to Dox and other chemotherapies, we performed a retrospective analysis of serum and plasma sphingolipids (SLs) from female patients with breast cancer (BC) undergoing anthracycline-containing therapy, correlating with cardiac parameters assessed by echocardiography. Results showed substantial changes in both plasma and serum SL species during therapy including ceramide (Cer), deoxydihydroCer, and dihydrosphingosine with reversion toward baseline after treatment. Linear mixed-effects model analysis revealed that baseline levels of a number of SLs correlated with adverse cardiac outcomes. Here, serum sphingosine-1-phosphate (S1P), dihydroS1P, and plasma Cer performed comparably to the prognostic value of pro-NT-BNP, an established biomarker of cardiotoxicity. Intriguingly, while pro-NT-BNP had no predictive value at mid- and post-therapy timepoints, serum S1P and dihydroS1P, and plasma Cer levels showed a correlation with adverse outcomes, particularly at the post-therapy timepoint. Finally, analysis of plasma and serum C16:C24-Cer ratios-previously linked with adverse cardiac outcomes-showed no correlation in the context of chemotherapy treatment. Overall, this pilot study provides initial evidence that plasma and serum SLs may have benefits as both prognostic and diagnostic biomarkers for female BC patients undergoing anthracycline-containing chemotherapy. Consequently, diagnostic SL measurements-recently implemented for metabolic-associated cardiac disorders-could have wider utility.
Collapse
Affiliation(s)
- Samia Mohammed
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | | | - Victoria Alvarado
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | | | - Christopher J Clarke
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Cancer Center, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
36
|
Wei J, Zhong Y, Li Y, Li J. Unraveling sphingolipid complexity in liver fibrosis: Perspectives for future research. Metabolism 2025:156254. [PMID: 40185396 DOI: 10.1016/j.metabol.2025.156254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Affiliation(s)
- Jiaqi Wei
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China; Southwest Medical University, Luzhou, China
| | - Yaohui Zhong
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China; Southwest Medical University, Luzhou, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China.
| | - Jun Li
- Southwest Medical University, Luzhou, China; Department of Anorectal, The Affiliated Hospital, Southwest Medical University, Luzhou. China.
| |
Collapse
|
37
|
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
|
38
|
Meshkovska Y, Dzhuraeva B, Godugu C, Pooladanda V, Thatikonda S. Deciphering the interplay: circulating cell-free DNA, signaling pathways, and disease progression in idiopathic pulmonary fibrosis. 3 Biotech 2025; 15:102. [PMID: 40165930 PMCID: PMC11954786 DOI: 10.1007/s13205-025-04272-y] [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: 06/12/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lung disease with an unknown etiology and a short survival rate. There is no accurate method of early diagnosis, and it involves computed tomography (CT) or lung biopsy. Since diagnostic methods are not accurate due to their similarity to other lung pathologies, discovering new biomarkers is a key issue for diagnosticians. Currently, the use of ccf-DNA (circulating cell-free deoxyribonucleic acid) is an important focus due to its association with IPF-induced alterations in metabolic pathways, such as amino acid metabolism, energy metabolism, and lipid metabolism pathways. Other biomarkers associated with metabolic changes have been found, and they are related to changes in type II/type I alveolar epithelial cells (AECs I/II), changes in extracellular matrix (ECM), and inflammatory processes. Currently, IPF pathogenetic treatment remains unknown, and the mortality rates are increasing, and the patients are diagnosed at a late stage. Signaling pathways and metabolic dysfunction have a significant role in the disease occurrence, particularly the transforming growth factor-β (TGF-β) signaling pathway, which plays an essential role. TGF-β, Wnt, Hedgehog (Hh), and integrin signaling are the main drivers of fibrosis. These pathways activate the transformation of fibroblasts into myofibroblasts, extracellular matrix (ECM) deposition, and tissue remodeling fibrosis. Therapy targeting diverse signaling pathways to slow disease progression is crucial in the treatment of IPF. Two antifibrotic medications, including pirfenidone and nintedanib, are Food and Drug Administration (FDA)-approved for treatment. ccf-DNA could become a new biomarker for IPF diagnosis to detect the disease at the early stage, while FDA-approved therapies could help to prevent late conditions from forming and decrease mortality rates.
Collapse
Affiliation(s)
- Yeva Meshkovska
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL 33612 USA
| | - Barchinai Dzhuraeva
- Department of Hospital Pediatrics, Moffitt Cancer Center, Tampa, FL 33612 USA
- Department of Hospital Pediatrics with a Course of Neonatology, National Center of Maternal and Child Health, Bishkek, 720017 Kyrgyzstan
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana 500037 India
| | - Venkatesh Pooladanda
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, 60 Blossom Street, Thier 9, Boston, MA 02114 USA
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, MA 02115 USA
| | - Sowjanya Thatikonda
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL 33612 USA
| |
Collapse
|
39
|
Blázquez AB, Mingo-Casas P, Quesada E, Priego EM, Pérez-Perez MJ, Martín-Acebes MA. Lipid-targeting antiviral strategies: Current state and future perspectives. Antiviral Res 2025; 236:106103. [PMID: 39947433 DOI: 10.1016/j.antiviral.2025.106103] [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: 12/11/2024] [Revised: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
There is an urgent need for antiviral compounds effective against currently known and future viral threats. The development of host-targeting antivirals (HTAs) appears as an alternative strategy to fight viral infections minimizing the potential of resistant mutant development and potentially leading to the identification of broad-spectrum antiviral agents. Among the host factors explored for HTA strategy, lipids constitute an attractive target as many viruses, even genetically diverse, hijack specific lipids during their lifecycle. Multiple repurposing efforts have been performed to analyze the antiviral properties of lipid-targeting compounds. These studies include the analysis of the effects of cholesterol lowering drugs such as statins, cholesterol transport inhibitors, sphingolipid modulators, de novo lipogenesis inhibitors blocking fatty acid synthesis, compounds targeting glycerophospholipids or drugs interfering with lipid droplet metabolism. This review is focused on the current status of lipid-based or lipid-targeting antiviral strategies and their potential for the development of antiviral therapies, with special emphasis on those studies that have reached advanced stages of development such as efficacy studies in animal models or clinical trials. Whereas there is still a long way to go, multiple proof-of-concept studies and clinical evidence reinforce the therapeutic potential of these strategies warranting their further development into effective antiviral therapies.
Collapse
Affiliation(s)
- Ana-Belén Blázquez
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid, Spain.
| | - Patricia Mingo-Casas
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid, Spain; Universidad Autónoma de Madrid (UAM, Escuela de Doctorado), Spain
| | | | | | | | - Miguel A Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid, Spain.
| |
Collapse
|
40
|
Fernàndez‐Bernal A, Sol J, Galo‐Licona JD, Mota‐Martorell N, Mas‐Bargues C, Belenguer‐Varea Á, Obis È, Viña J, Borrás C, Jové M, Pamplona R. Phenotypic upregulation of hexocylceramides and ether-linked phosphocholines as markers of human extreme longevity. Aging Cell 2025; 24:e14429. [PMID: 39639682 PMCID: PMC11984674 DOI: 10.1111/acel.14429] [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: 08/16/2024] [Revised: 10/14/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Centenarians and their relatives possess a notable survival advantage, with higher longevity and reduced susceptibility to major age-related diseases. To date, characteristic omics profiles of centenarians have been described, demonstrating that these individuals with exceptional longevity regulate their metabolism to adapt and incorporate more resilient biomolecules into their cells. Among these adaptations, the lipidomic profile stands out. However, it has not yet been determined whether this lipidomic profile is specific to centenarians or is the consequence of extreme longevity genetics and is also present in centenarians' offspring. This distinction is crucial for defining potential therapeutic targets that could help delay the aging process and associated pathologies. We applied mass-spectrometry-based techniques to quantify 569 lipid species in plasma samples from 39 centenarians, 63 centenarians' offspring, and 69 noncentenarians' offspring without familial connections. Based on this profile, we calculated different indexes to characterize the functional and structural properties of plasma lipidome. Our findings demonstrate that extreme longevity genetics (centenarians and centenarians' offspring) determines a specific lipidomic signature characterized by (i) an enrichment of hexosylceramides, (ii) a decrease of specific species of ceramides and sulfatides, (iii) a global increase of ether-PC and ether-LPC, and (iv) changes in the fluidity and diversity of specific lipid classes. We point out the conversion of ceramides to hexosylceramides and the maintenance of the levels of the ether-linked PC as a phenotypic trait to guarantee extreme longevity. We propose that this molecular signature is the result of an intrinsic adaptive program that preserves protective mechanisms and cellular identity.
Collapse
Affiliation(s)
- Anna Fernàndez‐Bernal
- Department of Experimental MedicineUniversity of Lleida‐Lleida Biomedical Research Institute (UdL‐IRBLleida)LleidaSpain
| | - Joaquim Sol
- Department of Experimental MedicineUniversity of Lleida‐Lleida Biomedical Research Institute (UdL‐IRBLleida)LleidaSpain
- Catalan Health Institute (ICS), Lleida Research Support Unit (USR)Fundació Institut Universitari per a la Recerca en Atenció Primària de Salut Jordi Gol i Gurina (IDIAP JGol)LleidaSpain
| | - José Daniel Galo‐Licona
- Department of Experimental MedicineUniversity of Lleida‐Lleida Biomedical Research Institute (UdL‐IRBLleida)LleidaSpain
| | - Natàlia Mota‐Martorell
- Department of Experimental MedicineUniversity of Lleida‐Lleida Biomedical Research Institute (UdL‐IRBLleida)LleidaSpain
| | - Cristina Mas‐Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable‐Instituto de Salud Carlos III (CIBERFES‐ISCIII)Institute of Health Research‐INCLIVA, University of ValenciaValènciaSpain
| | - Ángel Belenguer‐Varea
- Division of Geriatrics, Hospital Universitario de La Ribera (Alzira, Valencia, Spain), School of DoctorateUniversidad Católica de ValenciaValenciaSpain
| | - Èlia Obis
- Department of Experimental MedicineUniversity of Lleida‐Lleida Biomedical Research Institute (UdL‐IRBLleida)LleidaSpain
| | - José Viña
- Freshage Research Group, Department of Physiology, Faculty of Medicine, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable‐Instituto de Salud Carlos III (CIBERFES‐ISCIII)Institute of Health Research‐INCLIVA, University of ValenciaValènciaSpain
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable‐Instituto de Salud Carlos III (CIBERFES‐ISCIII)Institute of Health Research‐INCLIVA, University of ValenciaValènciaSpain
| | - Mariona Jové
- Department of Experimental MedicineUniversity of Lleida‐Lleida Biomedical Research Institute (UdL‐IRBLleida)LleidaSpain
| | - Reinald Pamplona
- Department of Experimental MedicineUniversity of Lleida‐Lleida Biomedical Research Institute (UdL‐IRBLleida)LleidaSpain
| |
Collapse
|
41
|
Choudhary P, Kumari S, Bagri K, Deshmukh R. Ceramide: a central regulator in Alzheimer's disease pathogenesis. Inflammopharmacology 2025; 33:1775-1783. [PMID: 40148603 DOI: 10.1007/s10787-025-01719-9] [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: 02/16/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025]
Abstract
Ceramide is a key component of sphingolipid metabolism and functions as a lipid second messenger. Sphingolipids are crucial for maintaining the nervous system, particularly in differentiation and development. Ceramide supports hippocampal growth but, at elevated levels, can impair dendritic cell function. During aging and neurodegenerative diseases like Alzheimer's disease (AD), intracellular ceramide production and accumulation increase, negatively impacting cognitive functions. High ceramide levels are linked to the progression of AD pathology, significantly contributing to amyloid β (Aβ) accumulation, tau tangle formation, insulin resistance, oxidative stress, and neuroinflammation. Ceramide facilitates the production and aggregation of Aβ peptides, leading to neurotoxic plaque formation. Its dysregulation is associated with abnormal tau protein phosphorylation, resulting in neurofibrillary tangles (NFTs). In addition, elevated ceramide levels can trigger brain inflammation by promoting the release of pro-inflammatory cytokines and activating microglia. This accumulation also enhances oxidative stress in neurons, damaging cellular components such as proteins, lipids, and DNA. This review will help in deeper understanding of the molecular pathways altered via ceramide metabolism and accumulation involved in the AD pathology. The cellular and pathological mechanisms of ceramide and their impact on Alzheimer's disease pathophysiology. A deeper understanding of ceramide-mediated effects in aging and AD could pave the way for innovative therapeutic strategies targeting ceramide metabolism to treat neurodegenerative diseases and age-related cognitive decline.
Collapse
Affiliation(s)
- Priyanka Choudhary
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, 151001, India
| | - Shilpa Kumari
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Kajal Bagri
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Rahul Deshmukh
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
42
|
Radermacher J, Erhardt VKJ, Walzer O, Haas EC, Kuppler KN, Zügner JSR, Lauer AA, Hartmann T, Grimm HS, Grimm MOW. Influence of Ibuprofen on glycerophospholipids and sphingolipids in context of Alzheimer´s Disease. Biomed Pharmacother 2025; 185:117969. [PMID: 40073745 DOI: 10.1016/j.biopha.2025.117969] [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: 08/28/2024] [Revised: 02/21/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is a multifactorial disorder associated with neuroinflammation, elevated oxidative stress, lipid alterations as well as amyloid-deposits and the formation of neurofibrillary tangles. Ibuprofen, a globally used analgesic, is discussed to influence disease progression due to its anti-inflammatory effect. However, changes in lipid-homeostasis induced by Ibuprofen have not yet been analyzed. Here we investigate the effect of Ibuprofen on lipid classes known to be associated with AD. Ibuprofen treatment leads to a significant increase in phosphatidylcholine, sphingomyelin and triacylglyceride (TAG) species whereas plasmalogens, which are highly susceptible for oxidation, were significantly decreased. The observed alterations in phosphatidylcholine and sphingomyelin levels in presence of Ibuprofen might counteract the reduced phosphatidylcholine- and sphingomyelin-levels found in AD brain tissue with potential positive aspects on synaptic plasticity and ceramide-induced apoptotic effects. On the other hand, Ibuprofen leads to elevated TAG-level resulting in the formation of lipid droplets which are associated with neuroinflammation. Reduction of plasmalogen-levels might accelerate decreased plasmalogen-levels found in AD brains. Treatment of Ibuprofen in terms of lipid-homeostasis reveals both potentially positive and negative changes relevant to AD. Therefore, understanding the influence of Ibuprofen on lipid-homeostasis may help to understand the heterogeneous results of studies treating AD with Ibuprofen.
Collapse
Affiliation(s)
| | | | - Oliver Walzer
- Experimental Neurology, Saarland University, Homburg, Saar 66424, Germany.
| | | | | | | | - Anna Andrea Lauer
- Experimental Neurology, Saarland University, Homburg, Saar 66424, Germany; Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, Leverkusen 51377, Germany.
| | - Tobias Hartmann
- Experimental Neurology, Saarland University, Homburg, Saar 66424, Germany; Deutsches Institut für Demenzprävention (DIDP), Saarland University, Homburg, Saar 66424, Germany.
| | - Heike Sabine Grimm
- Experimental Neurology, Saarland University, Homburg, Saar 66424, Germany; Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, Leverkusen 51377, Germany; Deutsches Institut für Demenzprävention (DIDP), Saarland University, Homburg, Saar 66424, Germany.
| | - Marcus Otto Walter Grimm
- Experimental Neurology, Saarland University, Homburg, Saar 66424, Germany; Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, Leverkusen 51377, Germany; Deutsches Institut für Demenzprävention (DIDP), Saarland University, Homburg, Saar 66424, Germany.
| |
Collapse
|
43
|
Mangla S, Ahlawat YK, Pathak G, Sharma N, Samani M, Bhan V, Essemine J, Sampasivam Y, Brar NS, Malik A, Ramteke V, Gupta S, Choubey S. Metabolic engineering of lipids for crop resilience and nutritional improvements towards sustainable agriculture. Funct Integr Genomics 2025; 25:78. [PMID: 40167787 DOI: 10.1007/s10142-025-01588-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/17/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Metabolic engineering of lipids in crops presents a promising strategy to enhance resilience against environmental stressors while improving nutritional quality. By manipulating key enzymes in lipid metabolism, introducing novel genes, and utilizing genome editing technologies, researchers have improved crop tolerance to abiotic stresses such as drought, salinity, and extreme temperatures. Additionally, modified lipid pathways contribute to resistance against biotic stresses, including pathogen attacks and pest infestations. Engineering multiple stress-resistance traits through lipid metabolism offers a holistic approach to strengthening crop resilience amid changing environmental conditions. Beyond stress tolerance, lipid engineering enhances the nutritional profile of crops by increasing beneficial lipids such as omega-3 fatty acids, vitamins, and antioxidants. This dual approach not only improves crop yield and quality but also supports global food security by ensuring sustainable agricultural production. Integrating advanced biotechnological tools with a deeper understanding of lipid biology paves the way for developing resilient, nutrient-rich crops capable of withstanding climate change and feeding a growing population.
Collapse
Affiliation(s)
- Swati Mangla
- Department of Biotechnology, University Institute of Engineering and Technology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Yogesh K Ahlawat
- Allied Health Sciences, Datta Meghe Institute of Higher Education and Research, Wardha, Maharashtra, India.
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India.
- Department of Biotechnology, University centre for research and development, Chandigarh University, Mohali, Punjab, India.
| | - Gaurav Pathak
- Department of Biotechnology, University Institute of Engineering and Technology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Nisha Sharma
- Department of Biotechnology, Graphic Era, Deemed to be University, Clement Town, Dehradun, 248002, Uttarakhand, India
| | - Maryam Samani
- Soil Sciences Department, University of Zanjan, Zanjan, Iran
| | - Veer Bhan
- Department of Biotechnology, University Institute of Engineering and Technology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Jemaa Essemine
- Bioinformatics Section, Department of Informatics, University of Quebec at Montreal, Pavillon President-Kennedy, Montreal, QC, H2X 3Y7, Canada
| | - Yashirdisai Sampasivam
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, UKM, 43600, Bangi, Selangor, Malaysia
| | - Navjot Singh Brar
- Department of Vegetable Sciences, Punjab Agricultural University, Ludhiana, Punjab, 144004, India
| | - Anurag Malik
- Division of Research and Innovation, Uttaranchal University, Dehradun, 24800, India
| | - Vikas Ramteke
- S. G. College of Agriculture and Research Station, Indira Gandhi Krishi Vishwavidyalaya, Jagdalpur, India
| | - Shivali Gupta
- Department of Chemistry, School of Chemical Engineering and Physical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Sumati Choubey
- Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab, India
| |
Collapse
|
44
|
Chitkara S, Atilla-Gokcumen GE. Decoding ceramide function: how localization shapes cellular fate and how to study it. Trends Biochem Sci 2025; 50:356-367. [PMID: 40000311 DOI: 10.1016/j.tibs.2025.01.007] [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: 07/01/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025]
Abstract
Recent studies emphasize that lipid synthesis, metabolism, and transport are crucial in modulating lipid function, underscoring the significance of lipid localization within the cell, in addition to their chemical structure. Ceramides stand out in this context because of their multifaceted roles in cellular processes. Here, we focus on the role of ceramides in apoptosis, senescence, and autophagy as these processes offer unique and contrasting perspectives on how ceramides function and can be intricately linked to their subcellular localization, providing critical insights into their complex biological interactions. Additionally, we highlight recent advancements in tools and techniques that have boosted our understanding of ceramide dynamics and different mechanisms of lipid functioning.
Collapse
Affiliation(s)
- Shweta Chitkara
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - G Ekin Atilla-Gokcumen
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA.
| |
Collapse
|
45
|
Zhang L, Ramesh P, Atencia Taboada L, Roessler R, Zijlmans DW, Vermeulen M, Picavet-Havik DI, van der Wel NN, Vaz FM, Medema JP. UGT8 mediated sulfatide synthesis modulates BAX localization and dictates apoptosis sensitivity of colorectal cancer. Cell Death Differ 2025; 32:657-671. [PMID: 39580596 PMCID: PMC11982410 DOI: 10.1038/s41418-024-01418-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
Elevated de novo lipid synthesis is a remarkable adaptation of cancer cells that can be exploited for therapy. However, the role of altered lipid metabolism in the regulation of apoptosis is still poorly understood. Using thermal proteome profiling, we identified Manidipine-2HCl, targeting UGT8, a key enzyme in the synthesis of sulfatides. In agreement, lipidomic analysis indicated that sulfatides are strongly reduced in colorectal cancer cells upon treatment with Manidipine-2HCl. Intriguingly, this reduction led to severe mitochondrial swelling and a strong synergism with BH3 mimetics targeting BCL-XL, leading to the activation of mitochondria-dependent apoptosis. Mechanistically, Manidipine-2HCl enhanced mitochondrial BAX localization in a sulfatide-dependent fashion, facilitating its activation by BH3 mimetics. In conclusion, our data indicates that UGT8 mediated synthesis of sulfatides controls mitochondrial homeostasis and BAX localization, dictating apoptosis sensitivity of colorectal cancer cells.
Collapse
Affiliation(s)
- Le Zhang
- LEXOR, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Prashanthi Ramesh
- LEXOR, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Lidia Atencia Taboada
- LEXOR, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Rebecca Roessler
- LEXOR, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Dick W Zijlmans
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
- Oncode Institute, Nijmegen, The Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
- Oncode Institute, Nijmegen, The Netherlands
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daisy I Picavet-Havik
- Medical Biology - MB Core Facility, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicole N van der Wel
- Medical Biology - MB Core Facility, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Frédéric M Vaz
- Amsterdam UMC location University of Amsterdam, Department of Laboratory Medicine and Pediatrics, Laboratory Genetic Metabolic Diseases, Emma Children's Hospital, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Inborn errors of metabolism, Amsterdam, The Netherlands
- Core Facility Metabolomics, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Paul Medema
- LEXOR, Center for Experimental Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Oncode Institute, Amsterdam, The Netherlands.
| |
Collapse
|
46
|
Huber K, Garg S, Schlautmann L, Wang R, He L, Huth R, Pouya A, Rohde C, Janssen M, Lüchtenborg C, Arnold C, Luque‐Navarro PM, Zaugg JB, Raffel S, Müller‐Tidow C, Jeremias I, López‐Cara LC, Brügger B, Pabst C. Phosphatidic acid phosphatase LPIN1 in phospholipid metabolism and stemness in hematopoiesis and AML. Hemasphere 2025; 9:e70118. [PMID: 40265168 PMCID: PMC12012646 DOI: 10.1002/hem3.70118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 12/14/2024] [Accepted: 02/12/2025] [Indexed: 04/24/2025] Open
Abstract
Targeting metabolism represents a promising approach to eradicate leukemic stem cells (LSCs) that are considered critical drivers of relapse in acute myeloid leukemia (AML). In this study, we demonstrate that the phosphatidic acid phosphatase LPIN1, which regulates the synthesis of diacylglycerol, the key substrate for triacylglycerol, and phospholipid production, is crucial for the function of healthy and leukemic hematopoietic stem and progenitor cells (HSPC and LSC). LPIN1 mRNA was highly expressed in the CD34+ compartment of primary human AML samples. LPIN1 suppression inhibited the proliferation of primary leukemic cells and normal HSPCs in vitro and in xenotransplantation assays. Lipidomics analyses revealed a reduction of phosphatidylcholine (PC) and phosphatidylethanolamine and an upregulation of sphingomyelin upon LPIN1 depletion. Distinct phospholipid composition was associated with genetic AML groups, and targeting PC production by choline kinase inhibitors showed strong anti-leukemic activity. In summary, our data establish a regulatory role of LPIN1 in HSPC and LSC function and provide novel insights into the role of glycerophospholipid homeostasis in stemness and differentiation.
Collapse
Affiliation(s)
- Karin Huber
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Department of Medicine II, Hematology and OncologyUniversity Hospital Schleswig HolsteinCampus KielGermany
| | - Swati Garg
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Department of Medical OncologyDana Farber Cancer InstituteBostonMassachusettsUSA
| | - Lena Schlautmann
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Rui Wang
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Lixiazi He
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Richard Huth
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Alireza Pouya
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Christian Rohde
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Maike Janssen
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | | | - Christian Arnold
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
- European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Pilar M. Luque‐Navarro
- Department of Pharmaceutical and Organic Chemistry, Faculty of PharmacyUniversity of GranadaGranadaSpain
| | - Judith B. Zaugg
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
- European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Simon Raffel
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Carsten Müller‐Tidow
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Irmela Jeremias
- Research Unit Apoptosis in Hematopoietic Stem CellsHelmholtz MunichOberschleißheimGermany
- Department of PediatricsDr. Von Hauner Children's Hospital, LMU University HospitalLMU MunichMunichGermany
- German Cancer Consortium (DKTK), Partner Site MunichMunichGermany
| | - Luisa C. López‐Cara
- Department of Pharmaceutical and Organic Chemistry, Faculty of PharmacyUniversity of GranadaGranadaSpain
| | - Britta Brügger
- Heidelberg University Biochemistry Center (BZH)HeidelbergGermany
| | - Caroline Pabst
- Department of Medicine V, Hematology, Oncology and RheumatologyUniversity Hospital HeidelbergHeidelbergGermany
- Molecular Medicine Partnership Unit (MMPU)University of Heidelberg and European Molecular Biology Laboratory (EMBL)HeidelbergGermany
| |
Collapse
|
47
|
Uzuner Odongo D, İlgün A, Bozkurt FB, Çakır T. A personalized metabolic modelling approach through integrated analysis of RNA-Seq-based genomic variants and gene expression levels in Alzheimer's disease. Commun Biol 2025; 8:502. [PMID: 40148444 PMCID: PMC11950204 DOI: 10.1038/s42003-025-07941-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Generating condition-specific metabolic models by mapping gene expression data to genome-scale metabolic models (GEMs) is a routine approach to elucidate disease mechanisms from a metabolic perspective. On the other hand, integrating variants that perturb enzyme functionality from the same RNA-seq data may enhance GEM accuracy, offering insights into genome-wide metabolic pathology. Our study pioneers the extraction of both transcriptomic and genomic data from the same RNA-seq data to reconstruct personalized metabolic models. We map genes with significantly higher load of pathogenic variants in Alzheimer's disease (AD) onto a human GEM together with the gene expression data. Comparative analysis of the resulting personalized patient metabolic models with the control models shows enhanced accuracy in detecting AD-associated metabolic pathways compared to the case where only expression data is mapped on the GEM. Besides, several otherwise would-be missed pathways are annotated in AD by considering the effect of genomic variants.
Collapse
Affiliation(s)
- Dilara Uzuner Odongo
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Atılay İlgün
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Fatma Betül Bozkurt
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, Gebze, Kocaeli, Turkey.
| |
Collapse
|
48
|
Liu X, Wang C, Sun X, Qiao Z, Yang X, Liu Y. Identify key transcript factors of adipocyte differentiation in abdominal fat of broilers based on ATAC-seq and RNA-seq. Poult Sci 2025; 104:105096. [PMID: 40157266 PMCID: PMC11995140 DOI: 10.1016/j.psj.2025.105096] [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: 12/03/2024] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Intensive breeding has resulted in excessive deposition of abdominal fat tissue (AFT) in broilers, leading to significant economic loss in the poultry industry. Understanding the molecular mechanisms underlying AFT development is essential for informed breeding strategies. In the current study, we elucidated dynamic changes of chromatin accessibility and transcriptional reprogramming in AFT at D14 and D42 in broilers based on integrated analysis of RNA-seq and ATAC-seq. RNA-seq analysis manifested significant transcriptional differences in AFT development, identifying 1323 up- and 1285 down-regulated differential expression genes (DEGs) as well as 63 up- and 58 down-regulated transcription factors (TFs) at D42 compared to those at D14. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of total DEGs revealed significant enrichment in pathways related to DNA replication, cell adhesion molecules, cell cycle, p53 signaling pathway, fatty acid degradation, fatty acid biosynthesis and steroid biosynthesis. Gene set enrichment analysis (GSEA) further indicated that autophagy, MAPK signaling pathway and inositol phosphate metabolism were up-regulated at D42 compared to D14, whereas cell cycle, DNA replication and steroid biosynthesis were down-regulated. Additionally, ATAC-seq analysis identified 394 gain and 1195 loss differentially accessible peaks (DPs) in AFT between D14 and D42, associated with 319 and 905 genes, respectively. These gain or loss genes were enriched in p53 signaling pathway, PPAR signaling pathway, fat digestion and absorption, FoxO signaling pathway and glycerol lipid metabolism. Integration analysis of ATAC-seq and RNA-seq data revealed 25 up-regulated and 75 down-regulated DEGs overlapping with genes linked to gain and loss DPs, respectively. Notably, ACACA, SCD, SREBF1and KLF9 exhibited significantly lower expression at D42 compared to D14. DNA motifs analysis identified NFIX and MYB as loss motifs, overlapping with down-regulated TFs, suggesting their potential role in AFT regulation. Furthermore, MYB and NFIX exhibited potential binding sites in the promoter regions of lipid metabolism-related genes (ELOVL6, PPARγ, FABP4, ACACA and SCD). Overall, these results will provide a theoretical basis for investigating the epigenetic modification and transcriptional regulation of AFT development in broilers.
Collapse
Affiliation(s)
- Xiaoying Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China.
| | - Chaohui Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China.
| | - Xi Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China.
| | - Zhihao Qiao
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China.
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China.
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, PR China.
| |
Collapse
|
49
|
Lee GB, Yang C, Hu F, Hao L. Evaluating sample normalization methods for MS-based multi-omics and the application to a neurodegenerative mouse model. Analyst 2025; 150:1271-1279. [PMID: 39995368 PMCID: PMC11851094 DOI: 10.1039/d4an01573h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Mass spectrometry (MS)-based omics methods have transformed biomedical research with accurate and high-throughput analysis of diverse molecules in biological systems. Recent technological advances also enabled multi-omics to be achieved from the same sample or on a single analytical platform. Sample normalization is a critical step in MS-omics studies but is usually conducted independently for each omics experiment. To bridge this technical gap, we evaluated different sample normalization methods suitable for analyzing proteins, lipids, and metabolites from the same sample for multi-omics analysis. We found that normalizing samples based on tissue weight or protein concentration before or after extraction generated distinct quantitative results. Normalizing samples first by tissue weight before extraction and then by protein concentration after extraction resulted in the lowest sample variation to reveal true biological differences. We then applied this two-step normalization method to investigate multi-omics profiles of mouse brains lacking the GRN gene. Loss-of-function mutations in the GRN gene lead to the deficiency of the progranulin protein and eventually cause neurodegeneration. Comparing the proteomics, lipidomics, and metabolomics profiles of GRN KO and WT mouse brains revealed molecular changes and pathways related to lysosomal dysfunction and neuroinflammation. In summary, we demonstrated the importance of selecting an appropriate normalization method during multi-omics sample preparation. Our normalization method is applicable to all tissue-based multi-omics studies, ensuring reliable and accurate biomolecule quantification for biological comparisons.
Collapse
Affiliation(s)
- Gwang Bin Lee
- Department of Chemistry & Biochemistry, University of Maryland, College Park, MD, 20742, USA.
| | - Cha Yang
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Ling Hao
- Department of Chemistry & Biochemistry, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
50
|
Zhang X, Chen J, Ma X, Tang X, Tan B, Liao P, Yao K, Jiang Q. Mycotoxins in Feed: Hazards, Toxicology, and Plant Extract-Based Remedies. Metabolites 2025; 15:219. [PMID: 40278348 PMCID: PMC12029259 DOI: 10.3390/metabo15040219] [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: 02/25/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
Background: Mycotoxins, which are secondary metabolites produced by fungi, are prevalent in animal feed and pose a serious risk to the healthy growth of livestock and poultry. Methods: This review aims to conclude current knowledge on the detrimental effects of mycotoxins on animal health and to demonstrate the potential of plant extracts as a means to counteract mycotoxin toxicity in feed. A systematic review of the literature was conducted to identify studies on the impact of mycotoxins on livestock and poultry health, as well as research into the use of plant extracts as feed additives to mitigate mycotoxin effects. Studies were selected based on their relevance to the topic, and data were extracted regarding the mechanisms of action and the efficacy of plant extracts. Results: Excessive mycotoxins in feed can lead to reduced appetite, impaired digestion, and general health issues in animals, resulting in decreased food intake, slowed weight gain, and instances of acute poisoning. Plant extracts with antioxidant, anti-inflammatory, and anti-mutagenic properties have shown the potential to improve production efficiency and reduce the toxic effects of mycotoxins. Conclusion: This comprehensive review not only consolidates the well-documented adverse effects of mycotoxins on animal health but also introduces a novel perspective by highlighting the potential of plant extracts as a promising and natural solution to counteract mycotoxin toxicity.
Collapse
Affiliation(s)
- Xiangnan Zhang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.Z.); (J.C.); (X.M.); (X.T.); (B.T.)
- Yuelushan Laboratory, Changsha 410128, China
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| | - Jiashun Chen
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.Z.); (J.C.); (X.M.); (X.T.); (B.T.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Xiaokang Ma
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.Z.); (J.C.); (X.M.); (X.T.); (B.T.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Xiongzhuo Tang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.Z.); (J.C.); (X.M.); (X.T.); (B.T.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Bie Tan
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.Z.); (J.C.); (X.M.); (X.T.); (B.T.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Peng Liao
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| | - Kang Yao
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| | - Qian Jiang
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.Z.); (J.C.); (X.M.); (X.T.); (B.T.)
- Yuelushan Laboratory, Changsha 410128, China
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China;
| |
Collapse
|