1
|
Potenza A, Gorla G, Carrozzini T, Pollaci G, Dei Cas M, Acerbi F, Vetrano IG, Ferroli P, Canavero I, Paroni R, Rifino N, Bersano A, Gatti L. Lipidomic profiling of the cerebrospinal fluid in moyamoya angiopathy patients. Orphanet J Rare Dis 2025; 20:243. [PMID: 40410905 PMCID: PMC12101001 DOI: 10.1186/s13023-025-03782-5] [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: 05/23/2024] [Accepted: 05/09/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND Moyamoya angiopathy (MA) is a rare cerebrovascular disorder which can occur in both children and young adults, characterized by progressive occlusion of the intracranial carotid arteries, leading patients to ischemic and haemorrhagic strokes. Despite decades of research, the mechanisms underlying MA remain poorly clarified and current gaps in the understanding of pathogenesis have hampered the development of suitable preventive strategies and therapeutic options. Moreover, clinically approved biomarkers for MA patients' stratification are missing. The unknown pathophysiology and the lack of reliable biomarkers prompted us to investigate cerebrospinal fluid (CSF) lipidome through state-of-the-art lipidomics. METHODS Intraoperative CSF from a subgroup of MA patients in comparison to age/sex matched controls (CTRL) was analysed through LC-MS/MS, by an untargeted lipidomic approach. Receiver operating characteristic (ROC) curve and simple linear regression analyses were performed for diagnostic use. We searched for simultaneously altered lipids in plasma and CSF of MA patients. RESULTS Overall, we observed a significant increase of sphingolipids (p < 0.05) and phospholipids (p < 0.05) in MA CSF. A partial least squares discriminant analysis clearly separated MA and CTRL by 64% on Principal Component 1. We identified lipid classes (n = 12) with a Variance Importance in Projection score ≥ 1.5, within those lipids highly correlated with MA (n = 70). A significant increase in acylcarnitines, sphingolipids (sphingomyelins and ceramides), phospholipids (lysophosphatidylcholines; phosphatidylcholines; phosphatidylethanolamines; ether-phosphatidylethanolamines; ether-phosphatidylcholines) and cholesterol esters was found by multivariate and univariate analyses. Monoacylglycerols were the only lipid class displaying a markedly significant (p < 0.001) decrease in CSF of MA patients as compared to CTRL subjects. The ROC curve and simple linear regression analysis identified 10 out of 12 lipid classes as reliable MA biomarkers, mainly dealing with phospholipids. We then compared current and previous data on plasma lipidomic profile. The discriminant analysis returned n = 175 (in plasma) and n = 70 (in CSF) simultaneously altered lipids respectively, and phosphatidylcholines (n = 10) resulted as commonly decreased in plasma and increased in CSF. CONCLUSIONS Our findings highlighted a strong pro-inflammatory environment in MA CSF. These preliminary hallmarks could be helpful to decipher the complex MA pathogenesis, by supplying candidate biomarkers for patient stratification.
Collapse
Affiliation(s)
- Antonella Potenza
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20122, Milan, Italy
| | - Gemma Gorla
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Tatiana Carrozzini
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Giuliana Pollaci
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20122, Milan, Italy
| | - Michele Dei Cas
- Clinical Biochemistry and Mass Spectrometry Lab, Department of Health Sciences, Università degli Studi di Milano, 20132, Milan, Italy
| | - Francesco Acerbi
- Neurosurgical Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126, Pisa, Italy
- Neurosurgery Unit, Pisa University Hospital, 56126, Pisa, Italy
| | - Ignazio G Vetrano
- Neurosurgical Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20122, Milan, Italy
| | - Paolo Ferroli
- Neurosurgical Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Isabella Canavero
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Rita Paroni
- Clinical Biochemistry and Mass Spectrometry Lab, Department of Health Sciences, Università degli Studi di Milano, 20132, Milan, Italy
| | - Nicola Rifino
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Anna Bersano
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Laura Gatti
- Laboratory of Neurobiology and UCV, Neurology IX Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy.
| |
Collapse
|
2
|
Ben X, Li C, Liu J, Liu T, Tong J, Li Q. Metabolomics-driven exploration of sphingosine 1-phosphate mechanisms in refractory epilepsy. Neurobiol Dis 2025; 212:106953. [PMID: 40349856 DOI: 10.1016/j.nbd.2025.106953] [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: 03/11/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025] Open
Abstract
This study aims to investigate the role of sphingosine 1-phosphate (S1P) in refractory epilepsy (RE) and elucidate its underlying molecular mechanisms. We employed metabolomics technology to analyze serum metabolites and gene expression patterns in individuals with RE. Additional omics analyses were conducted using cellular and animal models to explore the specific functions of S1P and related metabolic pathways. Our findings demonstrated that ACER3/SphK1/S1P play protective roles in maintaining mitochondrial structure and function. These elements were shown to mitigate neuronal hyperexcitability and protect against neuronal damage. By elucidating the dysregulation of metabolic pathways associated with disease onset and progression, our research illuminated the impact of abnormal sphingolipid metabolism and gene expression variances on the manifestation and progression of RE. This research underscores the critical impact of abnormal sphingolipid metabolism on RE development and progression. The insights gained from this study provide a foundation for developing targeted pharmaceutical interventions and symptomatic treatments for individuals with RE.
Collapse
Affiliation(s)
- Xinyu Ben
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Department of Neurology, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Chang Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Department of Neurology, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Jiaqi Liu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Department of Neurology, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Ting Liu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Department of Neurology, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China
| | - Jingyi Tong
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Department of Neurology, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China.
| | - Qifu Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, Department of Neurology, The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan 571199, China.
| |
Collapse
|
3
|
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
|
4
|
Guerra IMS, Rocha H, Moreira S, Gaspar A, Ferreira AC, Santos H, Rodrigues E, Castro-Chaves P, Melo T, Goracci L, Domingues P, Moreira ASP, Domingues MR. Lipidome plasticity in medium- and long-chain fatty acid oxidation disorders: Insights from dried blood spot lipidomics. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159621. [PMID: 40318842 DOI: 10.1016/j.bbalip.2025.159621] [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: 08/01/2024] [Revised: 04/14/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Fatty acid (FA) oxidation disorders (FAOD) are characterized by accumulation of specific acylcarnitines (CAR) and FA and can lead to potentially severe complications. In this study, dried blood spots (DBS) combined with LC-MS lipidomics analysis were used to assess lipidome plasticity in medium-chain acyl-CoA dehydrogenase deficiency (MCADD), long-chain hydroxyacyl-CoA dehydrogenase deficiency (LCHADD), and very long-chain acyl-CoA dehydrogenase deficiency (VLCADD), compared to control (CT) individuals, for screening potential prognostic biomarkers. Statistically significant variations were found in CAR, biomarkers for FAOD diagnosis, but other lipid species showed variations depending on the FAOD. Common changes in all FAOD included a few phosphatidylcholine (PC) lipid species, notably an up-regulation of LPC 16:1, possibly associated with a higher risk of cardiovascular disease (CVD). In LCHADD and VLCADD, an up-regulation of odd-chain PC (PC 33:0, PC 35:4 and PC 37:4) was observed. VLCADD exhibited higher levels of odd-chain TG, while LCHADD showed an up-regulation of ceramide (Cer 41:2;O2). The increase in the Cer class has been found to be associated with neurodegeneration and may contribute to the risk of developing this condition in LCHADD. An upregulation of ether-linked PC lipid species, including plasmenyl (known as endogenous antioxidants), was observed in MCADD, possibly as a response to increased oxidative stress reported in this disorder. Overall, DBS combined with lipidomics effectively pinpoints the lipid plasticity in FAOD, highlighting potential specific biomarkers for disease prognosis that warrant further validation for their association with the development of FAOD comorbidities.
Collapse
Affiliation(s)
- Inês M S Guerra
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Hugo Rocha
- Newborn Screening, Metabolism and Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, 4000-053 Porto, Portugal; Department of Pathological, Cytological and Thanatological Anatomy, School of Health, Polytechnic Institute of Porto, 4200-072 Porto, Portugal
| | - Sónia Moreira
- Reference Center for Hereditary Metabolic Diseases, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal; European Reference Network for Hereditary Metabolic Diseases - MetabERN, Portugal
| | - Ana Gaspar
- Inherited Metabolic Diseases Reference Centre, Lisboa Norte Hospital University Centre, Lisboa, Portugal
| | - Ana C Ferreira
- Inherited Metabolic Diseases Reference Center, Unidade Local de Saúde de São José, University Medical Centre of Lisbon, Portugal
| | - Helena Santos
- Inherited Metabolic Diseases Reference Centre, Vila Nova de Gaia Hospital Centre, Vila Nova de Gaia, Portugal
| | - Esmeralda Rodrigues
- Inherited Metabolic Diseases Reference Centre, São João Hospital University Centre, Porto, Portugal
| | - Paulo Castro-Chaves
- Inherited Metabolic Diseases Reference Centre, São João Hospital University Centre, Porto, Portugal
| | - Tânia Melo
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia (Perugia), Italy
| | - Pedro Domingues
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Ana S P Moreira
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - M Rosário Domingues
- CESAM - Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
5
|
Schindler RL, Jin LW, Zivkovic AM, Liu Y, Lebrilla CB. Region-specific quantitation of glycosphingolipids in the elderly human brain with Nanoflow MEA Chip Q/ToF mass spectrometry. Glycobiology 2025; 35:cwaf022. [PMID: 40207879 PMCID: PMC12021261 DOI: 10.1093/glycob/cwaf022] [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: 04/16/2024] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
Glycosphingolipids are a unique class of bioactive lipids responsible for lateral membrane organization and signaling found in high abundance in the central nervous system. Using nanoflow MEA Chip Q/ToF mass spectrometry, we profiled the intact glycosphingolipids of the elderly human brain in a region-specific manner. By chromatographic separation of glycan and ceramide isomers, we determined gangliosides to be the highest source of heterogeneity between regions with the expression of a- and b-series glycan structures. Investigation of these trends showed that specific glycan structures were, in part, determined by the structure of their lipid backbone. This study provides insight into the dynamic process of membrane remodeling in the brain during aging.
Collapse
Affiliation(s)
- Ryan L Schindler
- Department of Chemistry, University of California, Davis, CA 95616, United States
| | - Lee-way Jin
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, CA 95616, United States
| | - Angela M Zivkovic
- Department of Nutrition, University of California, Davis, CA 95616, United States
| | - Yiyun Liu
- Department of Chemistry, University of California, Davis, CA 95616, United States
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, CA 95616, United States
| |
Collapse
|
6
|
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
|
7
|
Shen X, Feng R, Zhou R, Zhang Z, Liu K, Wang S. Ceramide as a Promising Tool for Diagnosis and Treatment of Clinical Diseases: A Review of Recent Advances. Metabolites 2025; 15:195. [PMID: 40137159 PMCID: PMC11944470 DOI: 10.3390/metabo15030195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/27/2025] Open
Abstract
Background/Objectives: Ceramide, a sphingolipid metabolite, has emerged as a key player in various physiological and pathological processes. Changes in ceramide levels are associated with the occurrence and development of various diseases, highlighting its potential as a biomarker of various clinical diseases. Methods: The biosynthesis and metabolism of ceramide are discussed, along with its functions in cell signaling, apoptosis, and inflammation. This study further examines the potential of ceramide as a biomarker for disease diagnosis and treatment. Results: This article highlights the involvement of ceramide in several diseases, including cardiovascular diseases, dermatosis, cancer, neurodegenerative disorders and metabolic syndromes. For each disease, the potential of ceramide as a biomarker for disease diagnosis and prognosis is explored, and the feasibility of therapeutic strategies targeting ceramide metabolism are reviewed. Additionally, the challenges and future directions in the field of ceramide research are addressed. Conclusions: This review article provides an overview of the recent advances in understanding the role of ceramide in clinical diseases and its potential as a diagnostic and therapeutic tool.
Collapse
Affiliation(s)
- Xueping Shen
- School of Stomatology, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China;
| | - Rui Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Center for Scientific Research, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; (R.F.); (Z.Z.)
- School of Pharmacy, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China;
| | - Rui Zhou
- School of Pharmacy, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China;
| | - Zhaoyang Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Center for Scientific Research, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; (R.F.); (Z.Z.)
| | - Kaiyong Liu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei 230032, China
- Center for Big Data and Population Health, Institute of Health and Medicine, Hefei Comprehensive National Science Center, No 81 Meishan Road, Hefei 230032, China
- MOE Key Laboratory of Population Health Across Life Cycle, No. 81 Meishan Road, Hefei 230032, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, No. 81 Meishan Road, Hefei 230032, China
| | - Sheng Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Center for Scientific Research, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China; (R.F.); (Z.Z.)
- Center for Scientific Research, Anhui Medical University, No. 81 Meishan Road, Hefei 230032, China
| |
Collapse
|
8
|
Duan J, Sun J, Ma X, Du P, Dong P, Xue J, Lu Y, Jiang T. Association of escitalopram-induced shifts in gut microbiota and sphingolipid metabolism with depression-like behavior in wistar-kyoto rats. Transl Psychiatry 2025; 15:54. [PMID: 39962083 PMCID: PMC11833111 DOI: 10.1038/s41398-025-03277-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
The microbiota-gut-brain axis plays a pivotal role in neuropsychiatric disorders, particularly in depression. Escitalopram (ESC) is a first-line antidepressant, however, its regulatory mechanisms on the microbiota-gut-brain axis in the treatment of depression remain unclear. The antidepressant effects of ESC were evaluated using the forced swim test in Wistar-Kyoto (WKY) rats, while damage in the gut and brain regions was assessed through H&E staining and immunohistochemistry. The therapeutic mechanisms in WKY rats with depression-like behavior were investigated through 16S rRNA sequencing of the gut microbiota, serum untargeted metabolomics, and hippocampal proteomics. Results indicated that ESC intervention improved depressive-like behaviors, as evidenced by increased swimming times in WKY rats, and also restored intestinal permeability and brain tissue integrity. Significant changes in the gut microbiota composition, particularly an increase in Bacteroides barnesiae, as well as increases in serum sphingolipid metabolites (Sphinganine 1-phosphate, Sphingosine, and Sphingosine-1-phosphate) and hippocampal proteins (Sptlc1, Enpp5, Enpp2), were strongly correlated. These robust correlations suggest that ESC may exert its antidepressant effects by modulating sphingolipid metabolism through the influence of gut microbiota. Accordingly, this research elucidates novel mechanisms underlying the antidepressant efficacy of ESC and highlights the pivotal importance of the microbiota-gut-brain axis in mediating these effects.
Collapse
Affiliation(s)
- Jiajia Duan
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jiaxing Sun
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiao Ma
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Peipei Du
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Pengfei Dong
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Juan Xue
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yanli Lu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Tao Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
9
|
Niu H, Zhang M, Zhang K, Aishan S, Li H, Wu W. In-Depth Investigation on Potential Mechanism of Forest-Grown Ginseng Alleviating Alzheimer's Disease via UHPLC-MS-Based Metabolomics. Metabolites 2025; 15:93. [PMID: 39997718 PMCID: PMC11857256 DOI: 10.3390/metabo15020093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/24/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Alzheimer's disease is a central nervous system degenerative disease closely related to age with a complex pathogenesis. As a natural medicinal plant, forest-grown ginseng (GSF) contains abundant ginsenosides and offers significant neuroprotective effects. METHODS In this study, we comprehensively investigated the effect of GSF on the cell viability of PC12 cells in an AD model alongside metabolic changes in the serum and brains of mice, combined with an efficacy evaluation of PC12 cells in vitro and UHPLC-MS-based metabolomics in vivo. The goal of this study is to clarify the potential mechanism of GSF in treating AD. RESULTS The PC12 cell results showed that GSF can promote the proliferation of PC12 cells, reduce the content of IL-8, increase the activity of SOD, and alleviate the inflammation and oxidative stress induced by Aβ25~35. The immunohistochemical results for the mouse brain tissue also showed that GSF could reduce the inflammatory response of mouse brain tissue by reducing the overexpression of IBa1. AD was alleviated by reducing Aβ protein deposition in the mouse brain tissue. An untargeted metabolomics analysis was performed using UHPLC-Q-Exactive MS and principal component analysis (PCA) to identify the differentially expressed metabolites in the serum and brain tissue of AD mice after treatment. Twenty and seventeen different metabolites were identified in the serum and brain tissue, respectively. The pathway enrichment analysis of differential metabolites showed that GSF could treat AD by up-regulating succinic acid semialdehyde, carbamoyl phosphate, Sphingosine 1-phosphate, L-cystathionine, 2-ketobutyric acid, Vanillylmandelic acid, and D-Ribose to regulate sphingomyelin metabolism, the synthesis and metabolism of neurotransmitters and precursors, and energy metabolism. CONCLUSIONS GSF can reduce neuroinflammation and alleviate Alzheimer's disease by regulating the metabolic disorders of amino acids, sphingolipids, unsaturated fatty acids, and arachidonic acid in mice serum and brain tissue metabolites. These results suggest a link between metabolite imbalance and AD, and reveal the basis for the mechanism of ginsenosides in AD treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
10
|
He S, Xu Z, Han X. Lipidome disruption in Alzheimer's disease brain: detection, pathological mechanisms, and therapeutic implications. Mol Neurodegener 2025; 20:11. [PMID: 39871348 PMCID: PMC11773937 DOI: 10.1186/s13024-025-00803-6] [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: 08/19/2024] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
Alzheimer's disease (AD) is among the most devastating neurodegenerative disorders with limited treatment options. Emerging evidence points to the involvement of lipid dysregulation in the development of AD. Nevertheless, the precise lipidomic landscape and the mechanistic roles of lipids in disease pathology remain poorly understood. This review aims to highlight the significance of lipidomics and lipid-targeting approaches in the diagnosis and treatment of AD. We summarized the connection between lipid dysregulation in the human brain and AD at both genetic and lipid species levels. We briefly introduced lipidomics technologies and discussed potential challenges and areas of future advancements in the lipidomics field for AD research. To elucidate the central role of lipids in converging multiple pathological aspects of AD, we reviewed the current knowledge on the interplay between lipids and major AD features, including amyloid beta, tau, and neuroinflammation. Finally, we assessed the progresses and obstacles in lipid-based therapeutics and proposed potential strategies for leveraging lipidomics in the treatment of AD.
Collapse
Affiliation(s)
- Sijia He
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78299, USA
| | - Ziying Xu
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78299, USA.
| |
Collapse
|
11
|
Rufail ML, Bassi R, Giussani P. Sphingosine-1-Phosphate Metabolic Pathway in Cancer: Implications for Therapeutic Targets. Int J Mol Sci 2025; 26:1056. [PMID: 39940821 PMCID: PMC11817292 DOI: 10.3390/ijms26031056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Cancer biology revolves around understanding how cells undergo uncontrolled proliferation leading to the formation of malignant tumors. Key aspects include self-sufficiency in growth signals, the lack of response to signals of growth inhibition, the evasion of apoptosis, sustained angiogenesis, the evasion of immune response, the capacity to invade and metastasize, and alterations in cellular metabolism. A vast amount of research, which is exponentially growing, over the past few decades highlights the role of sphingolipids in cancer. They act not only as structural membrane components but also as bioactive molecules that regulate cell fate in different physio-pathological conditions. In cancer, sphingolipid metabolism is dysregulated, contributing to tumor progression, metastasis, and drug resistance. In this review, we outline the impact of sphingosine-1-phosphate (S1P) as a key bioactive sphingolipid in cancer. We give an overview of its metabolism summarizing the role of S1P as an intracellular and extracellular mediator through specific plasma membrane receptors in different cancers. We also describe previous findings on how the disruption in the balance between S1P and ceramide (Cer) is common in cancer cells and can contribute to tumorigenesis and resistance to chemotherapy. We finally consider the potential of targeting the metabolic pathways of S1P as well as its receptors and transporters as a promising therapeutic approach in cancer treatments.
Collapse
Affiliation(s)
- Miguel L. Rufail
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Rosaria Bassi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, LITA Segrate, Via Fratelli Cervi, 93, 20054 Segrate, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, LITA Segrate, Via Fratelli Cervi, 93, 20054 Segrate, Italy
| |
Collapse
|
12
|
Wang SX, Chen BS, Zhang ZJ, Zhu SR, Wang XL, Liu GQ. Isolation, structural elucidation and biosynthetic pathway of bioactive prenyl quinone compounds from Panus lecomtei based on untargeted metabolomics combined with molecular networking. Food Chem 2025; 463:141275. [PMID: 39298853 DOI: 10.1016/j.foodchem.2024.141275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Panus lecomtei is a relatively unfamiliar and undeveloped mushroom. This study generated ethyl acetate extracts of P. lecomtei intracellular (I), extracellular (E) and total fermentation broth (T). Both E and T extracts demonstrated antioxidant and antibacterial activities at 100 to 200 μg/mL. The composition differences of metabolites of these extracts were further studied based on comparative metabolomics by LS/MS and molecular network analysis. The results revealed that there were over 2000 significantly distinct metabolites among the three extracts, with abundant prenyl quinone compounds. Furthermore, the molecular network clarified the conversion relationship of P. lecomtei metabolites. Seven known prenyl quinone derivatives (1-7) were isolated from the E extract. Among them, compound 3 displayed excellent antioxidant activity and modest antibacterial activity. Compound 5 was discovered in fungi for the first time. Finally, a potential biosynthetic route for prenyl quinone in P. lecomtei was suggested.
Collapse
Affiliation(s)
- Si-Xian Wang
- Hunan Provincial Key Laboratory of Forestry Biotechnology & International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology, Central South University of Forestry & Technology, Changsha 410004, China
| | - Bao-Song Chen
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medicine School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zi-Juan Zhang
- Hunan Provincial Key Laboratory of Forestry Biotechnology & International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology, Central South University of Forestry & Technology, Changsha 410004, China
| | - Shi-Rong Zhu
- Hunan Provincial Key Laboratory of Forestry Biotechnology & International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology, Central South University of Forestry & Technology, Changsha 410004, China
| | - Xiao-Ling Wang
- Hunan Provincial Key Laboratory of Forestry Biotechnology & International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology, Central South University of Forestry & Technology, Changsha 410004, China.
| | - Gao-Qiang Liu
- Hunan Provincial Key Laboratory of Forestry Biotechnology & International Cooperation Base of Science and Technology Innovation on Forest Resource Biotechnology, Central South University of Forestry & Technology, Changsha 410004, China.
| |
Collapse
|
13
|
Wang S, Jin Z, Wu B, Morris AJ, Deng P. Role of dietary and nutritional interventions in ceramide-associated diseases. J Lipid Res 2025; 66:100726. [PMID: 39667580 PMCID: PMC11754522 DOI: 10.1016/j.jlr.2024.100726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024] Open
Abstract
Ceramides are important intermediates in sphingolipid metabolism and serve as signaling molecules with independent biological significance. Elevated cellular and circulating ceramide levels are consistently associated with pathological conditions including cardiometabolic diseases, neurological diseases, autoimmune diseases, and cancers. Although pharmacological inhibition of ceramide formation often protects against these diseases in animal models, pharmacological modulation of ceramides in humans remains impractical. Dietary interventions including the Mediterranean diet, lacto-ovo-vegetarian diet, calorie-restricted diet, restriction of dairy product consumption, and dietary supplementation with polyunsaturated fatty acids, dietary fibers, and polyphenols, all have beneficial effects on modulating ceramide levels. Mechanistic insights into these interventions are discussed. This article reviews the relationships between ceramides and disease pathogenesis, with a focus on dietary intervention as a viable strategy for lowering the concentration of circulating ceramides.
Collapse
Affiliation(s)
- Shengnan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Zihui Jin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Biyu Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Andrew J Morris
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Affairs Healthcare System, Little Rock, Arkansas, USA
| | - Pan Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
14
|
Fontana F, Donato AC, Malik A, Gelain F. Unveiling Interactions between Self-Assembling Peptides and Neuronal Membranes. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:26811-26823. [PMID: 39653368 DOI: 10.1021/acs.langmuir.4c02050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
The use of self-assembling peptide hydrogels in the treatment of spinal cord and brain injuries, especially when combined with adult neural stem cells, has shown great potential. To advance tissue engineering, it is essential to understand the effect of mechanochemical signaling on cellular differentiation. The elucidation of the molecular interactions at the level of the neuronal membrane still represents a promising area of investigation for many drug delivery and tissue engineering applications. An innovative molecular dynamics framework has been introduced to investigate the effect of SAP fibrils with different charges on neural membrane lipid domain dynamics. Such advance enables the in silico exploration of the biomimetic properties of SAP hydrogels and other polymeric biomaterials for tissue engineering applications.
Collapse
Affiliation(s)
- Federico Fontana
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unità di Ingegneria Tissutale, Viale Cappuccini 1, San Giovanni Rotondo, Foggia 71013, Italy
| | - Alice Cristina Donato
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Histology Unit, Department of Molecular Medicine, University of Padova, Padova 35121, Italy
| | - Ashish Malik
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unità di Ingegneria Tissutale, Viale Cappuccini 1, San Giovanni Rotondo, Foggia 71013, Italy
| | - Fabrizio Gelain
- Center for Nanomedicine and Tissue Engineering (CNTE), A.S.S.T. Grande Ospedale Metropolitano Niguarda, Piazza dell'Ospedale Maggiore 3, Milan 20162, Italy
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unità di Ingegneria Tissutale, Viale Cappuccini 1, San Giovanni Rotondo, Foggia 71013, Italy
| |
Collapse
|
15
|
Matthews DG, Khorani M, Bobe G, Caruso M, Magana AA, Gray NE, Quinn JF, Stevens JF, Maier CS, Soumyanath A. Centella asiatica improves cognitive function and alters the hippocampal metabolome of aged Tg2576 and wild-type mice. J Alzheimers Dis Rep 2024; 8:1611-1638. [PMID: 40034352 PMCID: PMC11863750 DOI: 10.1177/25424823241296740] [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: 10/04/2023] [Accepted: 09/23/2024] [Indexed: 03/05/2025] Open
Abstract
Background Alzheimer's disease (AD) is a growing public health problem in the aging population, with limited treatment options. We previously reported that Centella asiatica herb water extract (CAW) attenuates cognitive decline in murine models of AD and aging. Objective To explore changes in the hippocampal metabolome associated with CAW's modulation of cognitive function and amyloid-β (Aβ) plaque load in aged Tg2576 and wild-type (WT) mice. Methods We compared cognitive function, hippocampal Aβ plaque burden, and hippocampal metabolite profile in 20-month-old Tg2576 female mice and their WT littermates following 3-5 weeks treatment with CAW (0, 200, or 1000 mg/kg/d p.o.). Cognitive testing included contextual fear response (CFR) and novel object recognition task (NORT). Aβ plaque burden was measured via immunohistochemistry. Metabolomic profiles of mouse hippocampi were obtained using liquid chromatography coupled with high resolution tandem mass spectrometry. Results CAW treatment resulted in dose-related improvements in CFR and NORT performance of Tg2576 and WT mice. However, while CFR correlated with neurosignaling and glycosylated ceramide levels, NORT performance correlated with lysophosphatidylcholines and oxidized metabolites, and Aβ accumulation was linked to elevated excitatory and suppressed inhibitory neurotransmission. Only a subset of the metabolite changes induced by CAW in Tg2576 mice represented a reversal of metabolite differences between Tg2576 and WT mice, suggesting the involvement of other pathways in CAW's cognitive effects. Conclusions Mechanisms underlying CAW's cognitive effects extend beyond reversing metabolic effects of Aβ accumulation. The data support the potential use of CAW to manage memory challenges in aged individuals with or without AD.
Collapse
Affiliation(s)
- Donald G Matthews
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Mona Khorani
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | | | - Nora E Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, USA
- Parkinson's Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR, USA
| | - Jan F Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, USA
| | - Claudia S Maier
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
16
|
Lee W, Choi SH, Shea MG, Cheng P, Dombroski BA, Pitsillides AN, Heard‐Costa NL, Wang H, Bulekova K, Kuzma AB, Leung YY, Farrell JJ, Lin H, Kunkle BW, Naj A, Blue EE, Nusetor F, Wang D, Boerwinkle E, Bush WS, Zhang X, De Jager PL, Dupuis J, Farrer LA, Fornage M, Martin E, Pericak‐Vance M, Seshadri S, Wijsman EM, Wang L, The Alzheimer's Disease Sequencing Project, Schellenberg GD, Destefano AL, Haines JL, Peloso GM. Association of common and rare variants with Alzheimer's disease in more than 13,000 diverse individuals with whole-genome sequencing from the Alzheimer's Disease Sequencing Project. Alzheimers Dement 2024; 20:8470-8483. [PMID: 39428839 PMCID: PMC11667527 DOI: 10.1002/alz.14283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/08/2024] [Accepted: 09/05/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a common disorder of the elderly that is both highly heritable and genetically heterogeneous. METHODS We investigated the association of AD with both common variants and aggregates of rare coding and non-coding variants in 13,371 individuals of diverse ancestry with whole genome sequencing (WGS) data. RESULTS Pooled-population analyses of all individuals identified genetic variants at apolipoprotein E (APOE) and BIN1 associated with AD (p < 5 × 10-8). Subgroup-specific analyses identified a haplotype on chromosome 14 including PSEN1 associated with AD in Hispanics, further supported by aggregate testing of rare coding and non-coding variants in the region. Common variants in LINC00320 were observed associated with AD in Black individuals (p = 1.9 × 10-9). Finally, we observed rare non-coding variants in the promoter of TOMM40 distinct of APOE in pooled-population analyses (p = 7.2 × 10-8). DISCUSSION We observed that complementary pooled-population and subgroup-specific analyses offered unique insights into the genetic architecture of AD. HIGHLIGHTS We determine the association of genetic variants with Alzheimer's disease (AD) using 13,371 individuals of diverse ancestry with whole genome sequencing (WGS) data. We identified genetic variants at apolipoprotein E (APOE), BIN1, PSEN1, and LINC00320 associated with AD. We observed rare non-coding variants in the promoter of TOMM40 distinct of APOE.
Collapse
|
17
|
Martínez-Gardeazabal J, Pereira-Castelo G, Moreno-Rodríguez M, Llorente-Ovejero A, Fernández M, Fernández-Vega I, Manuel I, Rodríguez-Puertas R. Sphingosine 1-phosphate receptor subtype 1 (S1P 1) activity in the course of Alzheimer's disease. Neurobiol Dis 2024; 202:106713. [PMID: 39448041 DOI: 10.1016/j.nbd.2024.106713] [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: 07/15/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024] Open
Abstract
Some specific lipid molecules in the brain act as signaling molecules, neurotransmitters, or neuromodulators, by binding to specific G protein-coupled receptors (GPCR) for neurolipids. One such receptor, sphingosine 1-phosphate receptor subtype 1 (S1P1), is coupled to Gi/o proteins and is involved in cell proliferation, growth, and neuroprotection. S1P1 constitutes an interesting target for neurodegenerative diseases like multiple sclerosis and Alzheimer's disease (AD), in which changes in the sphingolipid metabolism have been observed. This study analyzes S1P1 receptor-mediated activity in healthy brains and during AD progression using postmortem samples from controls and patients at different Braak's stages. Additionally, the distribution of S1P1 receptor activity in human brains is compared to that in commonly used rodent models, rats and mice, through functional autoradiography, measuring [35S]GTPγS binding stimulated by the S1P1 receptor selective agonist CYM-5442 to obtain the distribution of functional activity of S1P1 receptors. S1P1 receptor-mediated activity, along with that of the cannabinoid CB1 receptor, is one of the highest recorded for any GPCR in many gray matter areas of the brain, reaching maximum values in the cerebellar cortex, specific areas of the hippocampus and the basal forebrain. S1P1 signaling is crucial in areas that regulate learning, memory, motor control, and nociception, such as the basal forebrain and basal ganglia. In AD, S1P1 receptor activity is increased in the inner layers of the frontal cortex and underlying cortical white matter at early stages, but decreases in the hippocampus in advanced stages, indicating ongoing brain impairment. Importantly, we identified significant correlations between S1P1 receptor activity and Braak stages, suggesting that S1P1 receptor dysfunction is associated to disease progression, particularly in memory-related regions. The S1P signaling via S1P1 receptor is a promising neurological target due to its role in key neurophysiological functions and its potential to modify the progression of neurodegenerative diseases. Finally, rats are suggested as a preferred experimental model for studying S1P1 receptor-mediated responses in the human brain.
Collapse
Affiliation(s)
- Jonatan Martínez-Gardeazabal
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain; Neurodegenerative Diseases, BioBizkaia Health Research Institute, Bizkaia, Spain, 48903 Barakaldo, Spain
| | - Gorka Pereira-Castelo
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Marta Moreno-Rodríguez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Alberto Llorente-Ovejero
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain
| | - Manuel Fernández
- Neurodegenerative Diseases, BioBizkaia Health Research Institute, Bizkaia, Spain, 48903 Barakaldo, Spain; Department of Neurology, Hospital Universitario de Cruces, 48903 Barakaldo, Spain
| | - Iván Fernández-Vega
- Department of Pathology, Hospital Universitario Central de Asturias, Avda. Roma, s/n, 33011 Oviedo, Spain; Health Research Institute of Principality of Asturias (ISPA), Av. del Hospital Universitario, s/n, 33011 Oviedo, Spain
| | - Iván Manuel
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain; Neurodegenerative Diseases, BioBizkaia Health Research Institute, Bizkaia, Spain, 48903 Barakaldo, Spain.
| | - Rafael Rodríguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), B° Sarriena s/n, 48940 Leioa, Spain; Neurodegenerative Diseases, BioBizkaia Health Research Institute, Bizkaia, Spain, 48903 Barakaldo, Spain
| |
Collapse
|
18
|
Limbu KR, Chhetri RB, Kim S, Shrestha J, Oh YS, Baek DJ, Park EY. Targeting sphingosine 1-phosphate and sphingosine kinases in pancreatic cancer: mechanisms and therapeutic potential. Cancer Cell Int 2024; 24:353. [PMID: 39462385 PMCID: PMC11514880 DOI: 10.1186/s12935-024-03535-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Pancreatic cancer is known to be the most lethal cancer. Fewer new treatments are being developed for pancreatic cancer as compared to other cancers. The bioactive lipid S1P, which is mainly regulated by sphingosine kinase 1 (SK1) and sphingosine kinase 2 (SK2) enzymes, plays significant roles in pancreatic cancer initiation and exacerbation. S1P controls many signaling pathways to modulate the progression of pancreatic cancer through the G-coupled receptor S1PR1-5. Several papers reporting amelioration of pancreatic cancer via modulation of S1P levels or downstream signaling pathways have previously been published. In this paper, for the first time, we have reviewed the results of previous studies to understand how S1P and its receptors contribute to the development of pancreatic cancer, and whether S1P can be a therapeutic target. In addition, we have also reviewed papers dealing with the effects of SK1 and SK2, which are kinases that regulate the level of S1P, on the pathogenesis of pancreatic cancer. We have also listed available drugs that particularly focus on S1P, S1PRs, SK1, and SK2 for the treatment of pancreatic cancer. Through this review, we would like to suggest that the SK/S1P/S1PR signaling system can be an important target for treating pancreatic cancer, where a new treatment target is desperately warranted.
Collapse
Affiliation(s)
- Khem Raj Limbu
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | | | - Subin Kim
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea
| | - Jitendra Shrestha
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam, 13135, South Korea
| | - Dong Jae Baek
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, Joennam, 58554, South Korea.
| |
Collapse
|
19
|
Shen H, Yu Y, Wang J, Nie Y, Tang Y, Qu M. Plasma lipidomic signatures of dementia with Lewy bodies revealed by machine learning, and compared to alzheimer's disease. Alzheimers Res Ther 2024; 16:226. [PMID: 39407312 PMCID: PMC11476188 DOI: 10.1186/s13195-024-01585-7] [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: 04/29/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Dementia with Lewy Bodies (DLB) is a complex neurodegenerative disorder that often overlaps clinically with Alzheimer's disease (AD), presenting challenges in accurate diagnosis and underscoring the need for novel biomarkers. Lipidomic emerges as a promising avenue for uncovering disease-specific metabolic alterations and potential biomarkers, particularly as the lipidomics landscape of DLB has not been previously explored. We aim to identify potential diagnostic biomarkers and elucidate the disease's pathophysiological mechanisms. METHODS This study conducted a lipidomic analysis of plasma samples from patients with DLB, AD, and healthy controls (HCs) at Xuanwu Hospital. Untargeted plasma lipidomic profiling was conducted via liquid chromatography coupled with mass spectrometry. Machine learning methods were employed to discern lipidomic signatures specific to DLB and to differentiate it from AD. RESULTS The study enrolled 159 participants, including 57 with AD, 48 with DLB, and 54 HCs. Significant differences in lipid profiles were observed between the DLB and HC groups, particularly in the classes of sphingolipids and phospholipids. A total of 55 differentially expressed lipid species were identified between DLB and HCs, and 17 between DLB and AD. Correlations were observed linking these lipidomic profiles to clinical parameters like Unified Parkinson's Disease Rating Scale III (UPDRS III) and cognitive scores. Machine learning models demonstrated to be highly effective in distinguishing DLB from both HCs and AD, achieving substantial accuracy through the utilization of specific lipidomic signatures. These include PC(15:0_18:2), PC(15:0_20:5), and SPH(d16:0) for differentiation between DLB and HCs; and a panel includes 13 lipid molecules: four PCs, two PEs, three SPHs, two Cers, and two Hex1Cers for distinguishing DLB from AD. CONCLUSIONS This study presents a novel and comprehensive lipidomic profile of DLB, distinguishing it from AD and HCs. Predominantly, sphingolipids (e.g., ceramides and SPHs) and phospholipids (e.g., PE and PC) were the most dysregulated lipids in relation to DLB patients. The lipidomics panels identified through machine learning may serve as effective plasma biomarkers for diagnosing DLB and differentiating it from AD dementia.
Collapse
Affiliation(s)
- Huixin Shen
- Departments of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yueyi Yu
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuting Nie
- Departments of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yi Tang
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Miao Qu
- Departments of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Departments of Chinese Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
20
|
Farooqui AA, Farooqui T. Phospholipids, Sphingolipids, and Cholesterol-Derived Lipid Mediators and Their Role in Neurological Disorders. Int J Mol Sci 2024; 25:10672. [PMID: 39409002 PMCID: PMC11476704 DOI: 10.3390/ijms251910672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Neural membranes are composed of phospholipids, sphingolipids, cholesterol, and proteins. In response to cell stimulation or injury, the metabolism of lipids generates various lipid mediators, which perform many cellular functions. Thus, phospholipids release arachidonic acid or docosahexaenoic acid from the sn-2 position of the glycerol moiety by the action of phospholipases A2. Arachidonic acid is a precursor for prostaglandins, leukotrienes, thromboxane, and lipoxins. Among these mediators, prostaglandins, leukotrienes, and thromboxane produce neuroinflammation. In contrast, lipoxins produce anti-inflammatory and pro-resolving effects. Prostaglandins, leukotrienes, and thromboxane are also involved in cell proliferation, differentiation, blood clotting, and blood vessel permeability. In contrast, DHA-derived lipid mediators are called specialized pro-resolving lipid metabolites (SPMs). They include resolvins, protectins, and maresins. These mediators regulate immune function by producing anti-inflammatory, pro-resolving, and cell protective effects. Sphingolipid-derived metabolites are ceramide, ceramide1-phosphate, sphingosine, and sphingosine 1 phosphate. They regulate many cellular processes, including enzyme activities, cell migration and adhesion, inflammation, and immunity. Cholesterol is metabolized into hydroxycholesterols and 7-ketocholesterol, which not only disrupts membrane fluidity, but also promotes inflammation, oxidative stress, and apoptosis. These processes lead to cellular damage.
Collapse
Affiliation(s)
| | - Tahira Farooqui
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA;
| |
Collapse
|
21
|
Bisht M, Kadian JP, Hooda T, Jain N, Lather A, Aggarwal N. Explore the Role of the Sphingosine-1-Phosphate Signalling as a Novel Promising Therapeutic Target for the Management of Parkinson's Disease. Drug Res (Stuttg) 2024; 74:365-378. [PMID: 39353579 DOI: 10.1055/a-2401-4578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Sphingosine-1-phosphate (S1P) is a cellular signalling molecule derived from sphingosine, which is a pro-apoptotic sphingolipid. Sphingolipids control various cellular actions like growth, homeostasis, and stress-related responses. The main sources of S1P in our body are erythrocytes. S1P controls both cellular mediators and other second messengers intracellularly. The S1P receptor also helps in inflammatory and neuroprotective effects (required to manage of Parkinson's). A large number of anti-Parkinson drugs are available, but still, there is a need for more effective and safer drugs. S1P and its receptors could be targeted as novel drugs due to their involvement in neuro-inflammation and Parkinson's. The present review effort to explore the biological role of S1P and related receptors, for their possible involvement in PD; furthermore. Overall, S1P and other related metabolizing enzymes have significant therapeutic opportunities for Parkinson's disease along with other neurological disorders.
Collapse
Affiliation(s)
- Manoj Bisht
- Devasthali Vidyapeeth College of Pharmacy, Rudrapur, Uttarakhand, India
| | - Jai Parkash Kadian
- Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh (Saharanpur)- UP India
| | - Tanuj Hooda
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Neelam Jain
- Department of Pharmaceutical Education & Research, Bhagat Phool Singh Mahila Vishwavidyalaya, Khanpur Kalan, Sonepat, Haryana, India
| | - Amit Lather
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| | - Navidha Aggarwal
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana, India
| |
Collapse
|
22
|
Liu J, Koutalos Y, Fan J. Lack of ceramide synthase 5 protects retinal ganglion cells from ocular hypertensive injury. Exp Eye Res 2024; 247:110061. [PMID: 39182597 PMCID: PMC11392625 DOI: 10.1016/j.exer.2024.110061] [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: 03/26/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
Ceramides with varying acyl-chain lengths can have unique biological actions and hence, cellular responses to ceramides may depend not on their overall concentration but on that of individual ceramide species. The purpose of this study was to determine individual ceramide species impacting retinal ganglion cell (RGC) loss under the ocular hypertensive condition. Induced pluripotent stem cell (iPSC)-derived RGCs and primary cultures of human astrocytes were used to determine the effect of individual ceramide species on both RGC viability and astrocyte secretion of inflammatory cytokines in vitro. In in vivo experiments with wild-type (WT) and ceramide synthase 5 (CerS5) knockout mice, intraocular pressure was unilaterally elevated with microbead injection. Retinal function and morphology were evaluated using pattern electroretinography (pERG) and immunofluorescence, respectively. Ceramide levels were determined by LC-MS/MS analysis. Exposure to C16:0-, C18:0-, C18:1-, C20:0- and C24:0-ceramides significantly reduces RGC viability in vitro, with the very long chain C24:0-ceramide being the most neurotoxic; treatment with C18:0-, C18:1- and C24:0-ceramides stimulates an increase of TNF-α secretion by astrocytes. The retinas of CerS5 KO mice have significantly reduced levels of C16:0- and C18:1-ceramides compared to WT; ocular hypertensive eyes of these mice maintain higher pERG amplitudes and RGC numbers compared to WT. Individual ceramides with different chain lengths have different effects on RGCs and astrocytes. Our results demonstrate that suppressing C16:0- and C18:1-ceramide species effectively protects RGCs against ocular hypertensive injury. These results provide a basis for targeting specific ceramide species in the treatment of glaucoma.
Collapse
Affiliation(s)
- Jian Liu
- Storm Eye Institute, Medical University of South Carolina, Department of Ophthalmology, 167 Ashley Ave, Charleston, SC, 29425, USA
| | - Yiannis Koutalos
- Storm Eye Institute, Medical University of South Carolina, Department of Ophthalmology, 167 Ashley Ave, Charleston, SC, 29425, USA
| | - Jie Fan
- Storm Eye Institute, Medical University of South Carolina, Department of Ophthalmology, 167 Ashley Ave, Charleston, SC, 29425, USA.
| |
Collapse
|
23
|
Yang C, Hu W, Liu J, Han C, Gao Q, Mei A, Zhou Y, Guo F, Han H. Achievements, challenges, and future prospects for industrialization of perovskite solar cells. LIGHT, SCIENCE & APPLICATIONS 2024; 13:227. [PMID: 39227394 PMCID: PMC11372181 DOI: 10.1038/s41377-024-01461-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 04/07/2024] [Accepted: 04/20/2024] [Indexed: 09/05/2024]
Abstract
In just over a decade, certified single-junction perovskite solar cells (PSCs) boast an impressive power conversion efficiency (PCE) of 26.1%. Such outstanding performance makes it highly viable for further development. Here, we have meticulously outlined challenges that arose during the industrialization of PSCs and proposed their corresponding solutions based on extensive research. We discussed the main challenges in this field including technological limitations, multi-scenario applications, sustainable development, etc. Mature photovoltaic solutions provide the perovskite community with invaluable insights for overcoming the challenges of industrialization. In the upcoming stages of PSCs advancement, it has become evident that addressing the challenges concerning long-term stability and sustainability is paramount. In this manner, we can facilitate a more effective integration of PSCs into our daily lives.
Collapse
Affiliation(s)
- Chuang Yang
- Michael Grätzel Center for Mesoscopic Solar Cells, Wuhan National Laboratory for Optoelectronics, Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Wenjing Hu
- Michael Grätzel Center for Mesoscopic Solar Cells, Wuhan National Laboratory for Optoelectronics, Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Jiale Liu
- Michael Grätzel Center for Mesoscopic Solar Cells, Wuhan National Laboratory for Optoelectronics, Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Chuanzhou Han
- Michael Grätzel Center for Mesoscopic Solar Cells, Wuhan National Laboratory for Optoelectronics, Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Qiaojiao Gao
- Michael Grätzel Center for Mesoscopic Solar Cells, Wuhan National Laboratory for Optoelectronics, Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Anyi Mei
- Michael Grätzel Center for Mesoscopic Solar Cells, Wuhan National Laboratory for Optoelectronics, Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Yinhua Zhou
- Michael Grätzel Center for Mesoscopic Solar Cells, Wuhan National Laboratory for Optoelectronics, Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Fengwan Guo
- Collaborative Innovation Center for Advanced Organic Chemical Materials, Co-constructed by the Province and Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062, Hubei, China.
| | - Hongwei Han
- Michael Grätzel Center for Mesoscopic Solar Cells, Wuhan National Laboratory for Optoelectronics, Key Laboratory of Materials Chemistry for Energy Conversion and Storage of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.
| |
Collapse
|
24
|
Acun AD, Kantar D. Modulation of oxidative stress and apoptosis by alteration of bioactive lipids in the pancreas, and effect of zinc chelation in a rat model of Alzheimer's disease. J Trace Elem Med Biol 2024; 85:127480. [PMID: 38875759 DOI: 10.1016/j.jtemb.2024.127480] [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: 03/13/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
INTRODUCTION Increasing epidemiological evidence highlights the association between systemic insulin resistance and Alzheimer's disease (AD). It is known that peripheral insulin resistance in the early stages of AD precedes and is a precursor to amyloid-β (Aβ) deposition. Although it is known that improving the CNS insulin sensitivity of AD patients is an important therapeutic goal and that the majority of insulin in the brain comes from the periphery, there has been little attention to the changes that occur in the pancreatic tissue of AD patients. Therefore, it is crucial to elucidate the mechanisms affecting insulin resistance in pancreatic tissue in AD. It is known that zinc (Zn2+) chelation is effective in reducing peripheral insulin resistance, cell apoptosis, cell death, and oxidative stress. OBJECTIVE It was aimed to determine the changes in bioactive lipids, amylin (AIPP), oxidative stress and apoptosis in pancreatic cells in the early stages of Alzheimer's disease. The main aim is to reveal the therapeutic effect of the Cyclo-Z agent on these changes seen in the pancreas due to AD disease. METHODS AD and ADC rats were intracerebroventricular (i.c.v.) Aβ1-42 oligomers. Cyclo-Z gavage was applied to ADC and SHC rats for 21 days. First of all, the effects of AIPP, bioactive ceramides, apoptosis and oxidative stress on the pancreatic tissue of AD group rats were evaluated. Then, the effect of Cyclo-Z treatment on these was examined. ELISA kit was used in biochemical analyses. RESULTS AIPP and ceramide (CER) levels and CER/ sphingosine-1 phosphate (S1P) ratio were increased in the pancreatic tissue of AD rats. It also increased the level of CER kinase (CERK), which is known to increase the concentration of CER 1-phosphate (C1P), which is known to be toxic to cells in the presence of excessive CER concentration. Due to the increase in CER level, it was observed that apoptosis and oxidative stress increased in the pancreatic cells of AD group rats. CONCLUSION Cyclo-Z, which has Zn2+ chelating properties, reduced AD model rats' AIPP level and oxidative stress and could prevent pancreatic apoptosis. Similar therapeutic effects were not observed in the pancreatic tissue of Cyclo-Z administered to the SH group. For this reason, it is thought that Cyclo-Z agent may have a therapeutic effect on the peripheral hyperinsulinemia observed in the early stages of AD disease and the resulting low amount of insulin transported to the brain, by protecting pancreatic cells from apoptosis and oxidative stress by regulating their bioactive metabolites.
Collapse
Affiliation(s)
- Alev Duygu Acun
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Arapsuyu, Antalya 07070, Turkey.
| | - Deniz Kantar
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Arapsuyu, Antalya 07070, Turkey
| |
Collapse
|
25
|
Zhang Z, Zhong Q, Qian Z, Zeng X, Zhang J, Xu X, Hylkema MN, Nolte IM, Snieder H, Huo X. Alterations of gut microbiota and its metabolomics in children with 6PPDQ, PBDE, PCB, and metal(loid) exposure. JOURNAL OF HAZARDOUS MATERIALS 2024; 475:134862. [PMID: 38885585 DOI: 10.1016/j.jhazmat.2024.134862] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024]
Abstract
The composition and metabolites of the gut microbiota can be altered by environmental pollutants. However, the effect of co-exposure to multiple pollutants on the human gut microbiota has not been sufficiently studied. In this study, gut microorganisms and their metabolites were compared between 33 children from Guiyu, an e-waste dismantling and recycling area, and 34 children from Haojiang, a healthy environment. The exposure level was assessed by estimating the daily intake (EDI) of polybrominated diphenyl ethers (PBDEs), polychlorinated biphenyls (PCBs), 6PPD-quinone (6PPDQ), and metal(loid)s in kindergarten dust. Significant correlations were found between the EDIs of 6PPDQ, BDE28, PCB52, Ni, Cu, and the composition of gut microbiota and specific metabolites. The Bayesian kernel machine regression model showed negative correlations between the EDIs of five pollutants (6PPDQ, BDE28, PCB52, Ni, and Cu) and the composition of gut microbiota. The EDIs of these five pollutants were positively correlated with the levels of the metabolite 2,4-diaminobutyric acid, while negatively correlated with the levels of d-erythro-sphingosine and d-threitol. Our study suggests that exposure to 6PPDQ, BDE28, PCB52, Ni, and Cu in kindergarten dust is associated with alterations in the composition and metabolites of the gut microbiota. These alterations may be associated with children's health.
Collapse
Affiliation(s)
- Zhuxia Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China
| | - Qi Zhong
- Laboratory of Environmental Medicine and Developmental Toxicology, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China; Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, Guangdong 510632, China
| | - Ziyi Qian
- Laboratory of Environmental Medicine and Developmental Toxicology, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China
| | - Xiang Zeng
- Laboratory of Environmental Medicine and Developmental Toxicology, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China; School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Jian Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Machteld N Hylkema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, the Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, the Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, the Netherlands
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, School of Environment, Jinan University, Guangzhou 511443, Guangdong, China; Laboratory of Environmental Medicine and Developmental Toxicology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong, China.
| |
Collapse
|
26
|
Chen CH, Cheng SN, Hu F, Su ZH, Wang KL, Cheng L, Chen J, Shi YR, Xia Y, Teng TY, Gao XY, Yavuz I, Lou YH, Wang ZK. Lead Isolation and Capture in Perovskite Photovoltaics toward Eco-Friendly Commercialization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403038. [PMID: 38724029 DOI: 10.1002/adma.202403038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/06/2024] [Indexed: 05/16/2024]
Abstract
Perovskite solar cells (PSCs) are developed rapidly in efficiency and stability in recent years, which can compete with silicon solar cells. However, an important obstacle to the commercialization of PSCs is the toxicity of lead ions (Pb2+) from water-soluble perovskites. The entry of free Pb2+ into organisms can cause severe harm to humans, such as blood lead poisoning, organ failure, etc. Therefore, this work reports a "lead isolation-capture" dual detoxification strategy with calcium disodium edetate (EDTA Na-Ca), which can inhibit lead leakage from PSCs under extreme conditions. More importantly, leaked lead exists in a nontoxic aggregation state chelated by EDTA. For the first time, in vivo experiments are conducted in mice to systematically prove that this material has a significant inhibitory effect on the toxicity of perovskites. In addition, this strategy can further enhance device performance, enabling the optimized devices to achieve an impressive power conversion efficiency (PCE) of 25.19%. This innovative strategy is a major breakthrough in the research on the prevention of lead toxicity in PSCs.
Collapse
Affiliation(s)
- Chun-Hao Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Advanced Negative Carbon Technologies, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Shu-Ning Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Advanced Negative Carbon Technologies, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Fan Hu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Advanced Negative Carbon Technologies, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Zhen-Huang Su
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201204, China
| | - Kai-Li Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Advanced Negative Carbon Technologies, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Liang Cheng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Advanced Negative Carbon Technologies, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Jing Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Advanced Negative Carbon Technologies, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yi-Ran Shi
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Advanced Negative Carbon Technologies, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Yu Xia
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Advanced Negative Carbon Technologies, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Tian-Yu Teng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Advanced Negative Carbon Technologies, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| | - Xing-Yu Gao
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201204, China
| | - Ilhan Yavuz
- Department of Physics, Marmara University, Ziverbey, Istanbul, 34722, Turkey
| | - Yan-Hui Lou
- College of Energy, Soochow Institute for Energy and Materials Innovations, Soochow University, Suzhou, 215006, China
| | - Zhao-Kui Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory of Advanced Negative Carbon Technologies, Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123, China
| |
Collapse
|
27
|
Iezhitsa I, Agarwal R, Agarwal P. Unveiling enigmatic essence of Sphingolipids: A promising avenue for glaucoma treatment. Vision Res 2024; 221:108434. [PMID: 38805893 DOI: 10.1016/j.visres.2024.108434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/05/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
Treatment of glaucoma, the leading cause of irreversible blindness, remains challenging. The apoptotic loss of retinal ganglion cells (RGCs) in glaucoma is the pathological hallmark. Current treatments often remain suboptimal as they aim to halt RGC loss secondary to reduction of intraocular pressure. The pathophysiological targets for exploring direct neuroprotective approaches, therefore are highly relevant. Sphingolipids have emerged as significant target molecules as they are not only the structural components of various cell constituents, but they also serve as signaling molecules that regulate molecular pathways involved in cell survival and death. Investigations have shown that a critical balance among various sphingolipid species, particularly the ceramide and sphingosine-1-phosphate play a role in deciding the fate of the cell. In this review we briefly discuss the metabolic interconversion of sphingolipid species to get an insight into "sphingolipid rheostat", the dynamic balance among metabolites. Further we highlight the role of sphingolipids in the key pathophysiological mechanisms that lead to glaucomatous loss of RGCs. Lastly, we summarize the potential drug candidates that have been investigated for their neuroprotective effects in glaucoma via their effects on sphingolipid axis.
Collapse
|
28
|
Minamihata T, Takano-Kawabe K, Moriyama M. Inhibition of Sphingosine Kinase 1 Reduces Sphingosine-1-Phosphate and Exacerbates Amyloid-Beta-Induced Neuronal Cell Death in Mixed-Glial-Cell Culture. Neurol Int 2024; 16:709-730. [PMID: 39051215 PMCID: PMC11270188 DOI: 10.3390/neurolint16040054] [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: 04/30/2024] [Revised: 06/24/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024] Open
Abstract
In Alzheimer's disease (AD) pathology, the accumulation of amyloid-beta (Aβ), a main component of senile plaques, activates glial cells and causes neuroinflammation. Excessive neuroinflammation results in neuronal dropouts and finally produces the symptoms of AD. Recent studies suggest that disorder in sphingosine-1-phosphate (S1P) metabolism, especially the decreased expression of sphingosine kinase (SK)1, followed by the reduction in the amount of S1P, can be a promotive factor in AD onset. Thus, we explored the possibility that dysregulated S1P metabolism affects AD through the altered function in glial cells. We evaluated the effect of PF-543, a pharmacological inhibitor of SK1, on the inflammatory responses by lipopolysaccharide (LPS)-activated glial cells, microglia, and astrocytes. The treatment with PF-543 decreased the intracellular S1P content in glial cells. The PF-543 treatment enhanced the nitric oxide (NO) production in the LPS-treated neuron/glia mixed culture. Furthermore, we found that the augmented production of NO and reactive oxygen species (ROS) in the PF-543-treated astrocytes affected the microglial inflammatory responses through humoral factors in the experiment using an astrocyte-conditioned medium. The PF-543 treatment also decreased the microglial Aβ uptake and increased the number of injured neurons in the Aβ-treated neuron/glia mixed culture. These results suggest that a decrease in the glial S1P content can exacerbate neuroinflammation and neurodegeneration through altered glial cell functions.
Collapse
Affiliation(s)
| | | | - Mitsuaki Moriyama
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, Izumisano 598-8531, Osaka, Japan; (T.M.); (K.T.-K.)
| |
Collapse
|
29
|
Faraji P, Kühn H, Ahmadian S. Multiple Roles of Apolipoprotein E4 in Oxidative Lipid Metabolism and Ferroptosis During the Pathogenesis of Alzheimer's Disease. J Mol Neurosci 2024; 74:62. [PMID: 38958788 PMCID: PMC11222241 DOI: 10.1007/s12031-024-02224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/14/2024] [Indexed: 07/04/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide and has a great socio-economic impact. Modified oxidative lipid metabolism and dysregulated iron homeostasis have been implicated in the pathogenesis of this disorder, but the detailed pathophysiological mechanisms still remain unclear. Apolipoprotein E (APOE) is a lipid-binding protein that occurs in large quantities in human blood plasma, and a polymorphism of the APOE gene locus has been identified as risk factors for AD. The human genome involves three major APOE alleles (APOE2, APOE3, APOE4), which encode for three subtly distinct apolipoprotein E isoforms (APOE2, APOE3, APOE4). The canonic function of these apolipoproteins is lipid transport in blood and brain, but APOE4 allele carriers have a much higher risk for AD. In fact, about 60% of clinically diagnosed AD patients carry at least one APOE4 allele in their genomes. Although the APOE4 protein has been implicated in pathophysiological key processes of AD, such as extracellular beta-amyloid (Aβ) aggregation, mitochondrial dysfunction, neuroinflammation, formation of neurofibrillary tangles, modified oxidative lipid metabolism, and ferroptotic cell death, the underlying molecular mechanisms are still not well understood. As for all mammalian cells, iron plays a crucial role in neuronal functions and dysregulation of iron homeostasis has also been implicated in the pathogenesis of AD. Imbalances in iron homeostasis and impairment of the hydroperoxy lipid-reducing capacity induce cellular dysfunction leading to neuronal ferroptosis. In this review, we summarize the current knowledge on APOE4-related oxidative lipid metabolism and the potential role of ferroptosis in the pathogenesis of AD. Pharmacological interference with these processes might offer innovative strategies for therapeutic interventions.
Collapse
Affiliation(s)
- Parisa Faraji
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Department of Biochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Hartmut Kühn
- Department of Biochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Shahin Ahmadian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
30
|
Thapa R, Moglad E, Afzal M, Gupta G, Bhat AA, Almalki WH, Kazmi I, Alzarea SI, Pant K, Ali H, Paudel KR, Dureja H, Singh TG, Singh SK, Dua K. ncRNAs and their impact on dopaminergic neurons: Autophagy pathways in Parkinson's disease. Ageing Res Rev 2024; 98:102327. [PMID: 38734148 DOI: 10.1016/j.arr.2024.102327] [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: 02/18/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Parkinson's Disease (PD) is a complex neurological illness that causes severe motor and non-motor symptoms due to a gradual loss of dopaminergic neurons in the substantia nigra. The aetiology of PD is influenced by a variety of genetic, environmental, and cellular variables. One important aspect of this pathophysiology is autophagy, a crucial cellular homeostasis process that breaks down and recycles cytoplasmic components. Recent advances in genomic technologies have unravelled a significant impact of ncRNAs on the regulation of autophagy pathways, thereby implicating their roles in PD onset and progression. They are members of a family of RNAs that include miRNAs, circRNA and lncRNAs that have been shown to play novel pleiotropic functions in the pathogenesis of PD by modulating the expression of genes linked to autophagic activities and dopaminergic neuron survival. This review aims to integrate the current genetic paradigms with the therapeutic prospect of autophagy-associated ncRNAs in PD. By synthesizing the findings of recent genetic studies, we underscore the importance of ncRNAs in the regulation of autophagy, how they are dysregulated in PD, and how they represent novel dimensions for therapeutic intervention. The therapeutic promise of targeting ncRNAs in PD is discussed, including the barriers that need to be overcome and future directions that must be embraced to funnel these ncRNA molecules for the treatment and management of PD.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Gaurav Gupta
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates; Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| | - Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Kumud Pant
- Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India; Graphic Era Hill University, Clement Town, Dehradun 248002, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Keshav Raj Paudel
- Centre of Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; School of Medical and Life Sciences, Sunway University, 47500 Sunway City, Malaysia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
31
|
Ebrahim N, Al Saihati HA, Alali Z, Aleniz FQ, Mahmoud SYM, Badr OA, Dessouky AA, Mostafa O, Hussien NI, Farid AS, El-Sherbiny M, Salim RF, Forsyth NR, Ali FEM, Alsabeelah NF. Exploring the molecular mechanisms of MSC-derived exosomes in Alzheimer's disease: Autophagy, insulin and the PI3K/Akt/mTOR signaling pathway. Biomed Pharmacother 2024; 176:116836. [PMID: 38850660 DOI: 10.1016/j.biopha.2024.116836] [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: 02/15/2024] [Revised: 05/16/2024] [Accepted: 05/26/2024] [Indexed: 06/10/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating neurological condition characterized by cognitive decline, motor coordination impairment, and amyloid plaque accumulation. The underlying molecular mechanisms involve oxidative stress, inflammation, and neuronal degeneration. This study aimed to investigate the therapeutic effects of mesenchymal stem cell-derived exosomes (MSC-exos) on AD and explore the molecular pathways involved, including the PI3K/Akt/mTOR axis, autophagy, and neuroinflammation. To assess the potential of MSC-exos for the treatment of AD, rats were treated with AlCl3 (17 mg/kg/once/day) for 8 weeks, followed by the administration of an autophagy activator (rapamycin), or MSC-exos with or without an autophagy inhibitor (3-methyladenin; 3-MA+ chloroquine) for 4 weeks. Memory impairment was tested, and brain tissues were collected for gene expression analyses, western blotting, histological studies, immunohistochemistry, and transmission electron microscopy. Remarkably, the administration of MSC-exos improved memory performance in AD rats and reduced the accumulation of amyloid-beta (Aβ) plaques and tau phosphorylation. Furthermore, MSC-exos promoted neurogenesis, enhanced synaptic function, and mitigated astrogliosis in AD brain tissues. These beneficial effects were associated with the modulation of autophagy and the PI3K/Akt/mTOR signalling pathway, as well as the inhibition of neuroinflammation. Additionally, MSC-exos were found to regulate specific microRNAs, including miRNA-21, miRNA-155, miRNA-17-5p, and miRNA-126-3p, further supporting their therapeutic potential. Histopathological and bioinformatic analyses confirmed these findings. This study provides compelling evidence that MSC-exos hold promise as a potential therapeutic approach for AD. By modulating the PI3K/Akt/mTOR axis, autophagy, and neuroinflammation, MSC-exos have the potential to improve memory, reduce Aβ accumulation, enhance neurogenesis, and mitigate astrogliosis. These findings shed light on the therapeutic potential of MSC-exos and highlight their role in combating AD.
Collapse
Affiliation(s)
- Nesrine Ebrahim
- Department of Medical Histology and Cell Biology Faculty of Medicine, Benha University, Benha, Egypt; Stem Cell Unit, Faculty of Medicine, Benha University, Egypt; Benha National University, Faculty of Medicine. student at Keele University, UK; Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Hajer A Al Saihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Albatin, Saudi Arabia; Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Zahraa Alali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, P.O Box 1803, Hafr Al Batin 31991, Saudi Arabia
| | - Faris Q Aleniz
- Department of Immunology, Collage of Applied Science, Alkharj
| | - Sabry Younis Mohamed Mahmoud
- Biology Department, College of Sciences, University of Hafr Al Batin, P. O. Box 1803, Hafar Al Batin 31991, Saudi Arabia. Agricultural Microbiology Department, Faculty of Agriculture, Sohag University, Sohag, Egypt
| | - Omnia A Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Benha, Egypt
| | - Arigue A Dessouky
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Egypt
| | - Ola Mostafa
- Department of Medical Histology and Cell Biology Faculty of Medicine, Benha University, Benha, Egypt
| | - Noha I Hussien
- Department of Physiology, Faculty of Medicine, Benha University, Egypt
| | - Ayman Samir Farid
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Qalyubia 13736, Egypt
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Rabab F Salim
- Department of Medical Biochemistry and molecular biology, Faculty of Medicine, Benha University, Egypt
| | - Nicholas Robert Forsyth
- School of Pharmacy and Bioengineering, Keele University. Vice Principals' Office, University of Aberdeen, Kings College, Aberdeen, AB24 3FX, UK
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| | - Nimer F Alsabeelah
- Assistant Professor of Pharmacology Pharmacy Practice Department, Pharmacy College University of Hafr Al Batin, P.O. Box 1803, Hafr Al Batin 31991, Saudi Arabia
| |
Collapse
|
32
|
Sanluca C, Spagnolo P, Mancinelli R, De Bartolo MI, Fava M, Maccarrone M, Carotti S, Gaudio E, Leuti A, Vivacqua G. Interaction between α-Synuclein and Bioactive Lipids: Neurodegeneration, Disease Biomarkers and Emerging Therapies. Metabolites 2024; 14:352. [PMID: 39057675 PMCID: PMC11278689 DOI: 10.3390/metabo14070352] [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: 05/12/2024] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
The present review provides a comprehensive examination of the intricate dynamics between α-synuclein, a protein crucially involved in the pathogenesis of several neurodegenerative diseases, including Parkinson's disease and multiple system atrophy, and endogenously-produced bioactive lipids, which play a pivotal role in neuroinflammation and neurodegeneration. The interaction of α-synuclein with bioactive lipids is emerging as a critical factor in the development and progression of neurodegenerative and neuroinflammatory diseases, offering new insights into disease mechanisms and novel perspectives in the identification of potential biomarkers and therapeutic targets. We delve into the molecular pathways through which α-synuclein interacts with biological membranes and bioactive lipids, influencing the aggregation of α-synuclein and triggering neuroinflammatory responses, highlighting the potential of bioactive lipids as biomarkers for early disease detection and progression monitoring. Moreover, we explore innovative therapeutic strategies aimed at modulating the interaction between α-synuclein and bioactive lipids, including the development of small molecules and nutritional interventions. Finally, the review addresses the significance of the gut-to-brain axis in mediating the effects of bioactive lipids on α-synuclein pathology and discusses the role of altered gut lipid metabolism and microbiota composition in neuroinflammation and neurodegeneration. The present review aims to underscore the potential of targeting α-synuclein-lipid interactions as a multifaceted approach for the detection and treatment of neurodegenerative and neuroinflammatory diseases.
Collapse
Affiliation(s)
- Chiara Sanluca
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Paolo Spagnolo
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Romina Mancinelli
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, 00185 Rome, Italy (E.G.)
| | | | - Marina Fava
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
| | - Mauro Maccarrone
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Simone Carotti
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
| | - Eugenio Gaudio
- Department of Anatomic, Histologic, Forensic and Locomotor Apparatus Sciences, Sapienza University of Roma, 00185 Rome, Italy (E.G.)
| | - Alessandro Leuti
- Biochemistry and Molecular Biology Unit, Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy;
| | - Giorgio Vivacqua
- Department of Medicine, Laboratory of Microscopic and Ultrastructural Anatomy, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy (S.C.)
| |
Collapse
|
33
|
Tong B, Ba Y, Li Z, Yang C, Su K, Qi H, Zhang D, Liu X, Wu Y, Chen Y, Ling J, Zhang J, Yin X, Yu P. Targeting dysregulated lipid metabolism for the treatment of Alzheimer's disease and Parkinson's disease: Current advancements and future prospects. Neurobiol Dis 2024; 196:106505. [PMID: 38642715 DOI: 10.1016/j.nbd.2024.106505] [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: 01/28/2024] [Revised: 03/02/2024] [Accepted: 04/14/2024] [Indexed: 04/22/2024] Open
Abstract
Alzheimer's and Parkinson's diseases are two of the most frequent neurological diseases. The clinical features of AD are memory decline and cognitive dysfunction, while PD mainly manifests as motor dysfunction such as limb tremors, muscle rigidity abnormalities, and slow gait. Abnormalities in cholesterol, sphingolipid, and glycerophospholipid metabolism have been demonstrated to directly exacerbate the progression of AD by stimulating Aβ deposition and tau protein tangles. Indirectly, abnormal lipids can increase the burden on brain vasculature, induce insulin resistance, and affect the structure of neuronal cell membranes. Abnormal lipid metabolism leads to PD through inducing accumulation of α-syn, dysfunction of mitochondria and endoplasmic reticulum, and ferroptosis. Great progress has been made in targeting lipid metabolism abnormalities for the treatment of AD and PD in recent years, like metformin, insulin, peroxisome proliferator-activated receptors (PPARs) agonists, and monoclonal antibodies targeting apolipoprotein E (ApoE). This review comprehensively summarizes the involvement of dysregulated lipid metabolism in the pathogenesis of AD and PD, the application of Lipid Monitoring, and emerging lipid regulatory drug targets. A better understanding of the lipidological bases of AD and PD may pave the way for developing effective prevention and treatment methods for neurodegenerative disorders.
Collapse
Affiliation(s)
- Bin Tong
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China; School of Ophthalmology and Optometry of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Yaoqi Ba
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China; School of Ophthalmology and Optometry of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Zhengyang Li
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China; The First Clinical Medical College of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Caidi Yang
- The First Clinical Medical College of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Kangtai Su
- The First Clinical Medical College of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Haodong Qi
- The First Clinical Medical College of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Deju Zhang
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China; Center for Clinical Precision Medicine, Jiujiang University, Jiujiang, China; Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Xiao Liu
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China; Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuting Wu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Jitao Ling
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China
| | - Jing Zhang
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China; Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China.
| | - Xiaoping Yin
- Department of Neurology, Affiliated Hospital of Jiujiang University, Jiujiang, China; Center for Clinical Precision Medicine, Jiujiang University, Jiujiang, China.
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China.
| |
Collapse
|
34
|
Galatolo D, Rocchiccioli S, Di Giorgi N, Dal Canto F, Signore G, Morani F, Ceccherini E, Doccini S, Santorelli FM. Proteomics and lipidomic analysis reveal dysregulated pathways associated with loss of sacsin. Front Neurosci 2024; 18:1375299. [PMID: 38911600 PMCID: PMC11191878 DOI: 10.3389/fnins.2024.1375299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Introduction Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is a rare incurable neurodegenerative disease caused by mutations in the SACS gene, which codes for sacsin, a large protein involved in protein homeostasis, mitochondrial function, cytoskeletal dynamics, autophagy, cell adhesion and vesicle trafficking. However, the pathogenic mechanisms underlying sacsin dysfunction are still largely uncharacterized, and so attempts to develop therapies are still in the early stages. Methods To achieve further understanding of how processes are altered by loss of sacsin, we used untargeted proteomics to compare protein profiles in ARSACS fibroblasts versus controls. Results Our analyses confirmed the involvement of known biological pathways and also implicated calcium and lipid homeostasis in ARSACS skin fibroblasts, a finding further verified in SH-SY5Y SACS -/- cells. Validation through mass spectrometry-based analysis and comparative quantification of lipids by LC-MS in fibroblasts revealed increased levels of ceramides coupled with a reduction of diacylglycerols. Discussion In addition to confirming aberrant Ca2+ homeostasis in ARSACS, this study described abnormal lipid levels associated with loss of sacsin.
Collapse
Affiliation(s)
| | | | | | | | - Giovanni Signore
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
- Department of Biology, University of Pisa, Pisa, Italy
| | - Federica Morani
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Pisa, Italy
| | - Elisa Ceccherini
- Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Stefano Doccini
- Molecular Medicine, IRCCS Stella Maris Foundation, Pisa, Italy
| | | |
Collapse
|
35
|
Zhang S, Tang S, Liu Z, Lv H, Cai X, Zhong R, Chen L, Zhang H. Baicalin restore intestinal damage after early-life antibiotic therapy: the role of the MAPK signaling pathway. Pharmacol Res 2024; 204:107194. [PMID: 38663526 DOI: 10.1016/j.phrs.2024.107194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 04/30/2024]
Abstract
Antibiotic related intestinal injury in early life affects subsequent health and susceptibility. Here, we employed weaned piglets as a model to investigate the protective effects of baicalin against early-life antibiotic exposure-induced microbial dysbiosis. Piglets exposed to lincomycin showed a marked reduction in body weight (p < 0.05) and deterioration of jejunum intestinal morphology, alongside an increase in antibiotic-resistant bacteria such as Staphylococcus, Dolosicoccus, Escherichia-Shigella, and Raoultella. In contrast, baicalin treatment resulted in body weights, intestinal morphology, and microbial profiles that closely resembled those of the control group (p > 0.05), with a significant increase in norank_f_Muribaculaceae and Prevotellaceae_NK3B31_group colonization compared with lincomycin group (p < 0.05). Further analysis through fecal microbial transplantation into mice revealed that lincomycin exposure led to significant alterations in intestinal morphology and microbial composition, notably increasing harmful microbes and decreasing beneficial ones such as norank_Muribaculaceae and Akkermansia (p < 0.05). This shift was associated with an increase in harmful metabolites and disruption of the calcium signaling pathway gene expression. Conversely, baicalin supplementation not only counteracted these effects but also enhanced beneficial metabolites and regulated genes within the MAPK signaling pathway (MAP3K11, MAP4K2, MAPK7, MAPK13) and calcium channel proteins (ORA13, CACNA1S, CACNA1F and CACNG8), suggesting a mechanism through which baicalin mitigates antibiotic-induced intestinal and microbial disturbances. These findings highlight baicalin's potential as a plant extract-based intervention for preventing antibiotic-related intestinal injury and offer new targets for therapeutic strategies.
Collapse
Affiliation(s)
- Shunfen Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shanlong Tang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhengqun Liu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Tianjin Key Laboratory of Animal Molecular Breeding and Biotechnology, Tianjin Engineering Research Center of Animal Healthy Farming, Institute of Animal Science and Veterinary, Tianjin Academy of Agricultural Sciences, Tianjin 300381, China
| | - Huiyuan Lv
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Beijing Centre Biology Co., Ltd., Daxing District, Beijing 102218, China
| | - Xueying Cai
- Department of Critical Care, Hangzhou First People's Hospital, Hangzhou 310003, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Liang Chen
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
36
|
Zhang YQ, Li J, Qin Z, Li DM, Ye FZ, Bei SH, Zhang XH, Feng L. METTL5 promotes gastric cancer progression via sphingomyelin metabolism. World J Gastrointest Oncol 2024; 16:1925-1946. [PMID: 38764837 PMCID: PMC11099429 DOI: 10.4251/wjgo.v16.i5.1925] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/09/2024] [Accepted: 02/19/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND The treatment of gastric cancer (GC) has caused an enormous social burden worldwide. Accumulating studies have reported that N6-methyladenosine (m6A) is closely related to tumor progression. METTL5 is a m6A methyltransferase that plays a pivotal role in maintaining the metabolic stability of cells. However, its aberrant regulation in GC has not been fully elucidated. AIM To excavate the role of METTL5 in the development of GC. METHODS METTL5 expression and clinicopathological characteristics were analyzed via The Cancer Genome Atlas dataset and further verified via immunohistochemistry, western blotting and real-time quantitative polymerase chain reaction in tissue microarrays and clinical samples. The tumor-promoting effect of METTL5 on HGC-27 and AGS cells was explored in vitro by Cell Counting Kit-8 assays, colony formation assays, scratch healing assays, transwell assays and flow cytometry. The tumor-promoting role of METTL5 in vivo was evaluated in a xenograft tumor model. The EpiQuik m6A RNA Methylation Quantification Kit was used for m6A quantification. Next, liquid chromatography-mass spectrometry was used to evaluate the association between METTL5 and sphingomyelin metabolism, which was confirmed by Enzyme-linked immunosorbent assay and rescue tests. In addition, we investigated whether METTL5 affects the sensitivity of GC cells to cisplatin via colony formation and transwell experiments. RESULTS Our research revealed substantial upregulation of METTL5, which suggested a poor prognosis of GC patients. Increased METTL5 expression indicated distant lymph node metastasis, advanced cancer stage and pathological grade. An increased level of METTL5 correlated with a high degree of m6A methylation. METTL5 markedly promotes the proliferation, migration, and invasion of GC cells in vitro. METTL5 also promotes the growth of GC in animal models. METTL5 knockdown resulted in significant changes in sphingomyelin metabolism, which implies that METTL5 may impact the development of GC via sphingomyelin metabolism. In addition, high METTL5 expression led to cisplatin resistance. CONCLUSION METTL5 was found to be an oncogenic driver of GC and may be a new target for therapy since it facilitates GC carcinogenesis through sphingomyelin metabolism and cisplatin resistance.
Collapse
Affiliation(s)
- Ya-Qiong Zhang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Jian Li
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Zhe Qin
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - De-Ming Li
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Fang-Zhou Ye
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Song-Hua Bei
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Xiao-Hong Zhang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| |
Collapse
|
37
|
Poinsot V, Pizzinat N, Ong-Meang V. Engineered and Mimicked Extracellular Nanovesicles for Therapeutic Delivery. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:639. [PMID: 38607173 PMCID: PMC11013861 DOI: 10.3390/nano14070639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Exosomes are spherical extracellular nanovesicles with an endosomal origin and unilamellar lipid-bilayer structure with sizes ranging from 30 to 100 nm. They contain a large range of proteins, lipids, and nucleic acid species, depending on the state and origin of the extracellular vesicle (EV)-secreting cell. EVs' function is to encapsulate part of the EV-producing cell content, to transport it through biological fluids to a targeted recipient, and to deliver their cargos specifically within the aimed recipient cells. Therefore, exosomes are considered to be potential biological drug-delivery systems that can stably deliver their cargo into targeted cells. Various cell-derived exosomes are produced for medical issues, but their use for therapeutic purposes still faces several problems. Some of these difficulties can be avoided by resorting to hemisynthetic approaches. We highlight here the uses of alternative exosome-mimes involving cell-membrane coatings on artificial nanocarriers or the hybridization between exosomes and liposomes. We also detail the drug-loading strategies deployed to make them drug-carrier systems and summarize the ongoing clinical trials involving exosomes or exosome-like structures. Finally, we summarize the open questions before considering exosome-like disposals for confident therapeutic delivery.
Collapse
Affiliation(s)
- Verena Poinsot
- Inserm, CNRS, Faculté de Santé, Université Toulouse III—Paul Sabatier, I2MC U1297, 31432 Toulouse, France; (N.P.); (V.O.-M.)
| | | | | |
Collapse
|
38
|
Zhang X, Chen C, Liu Y. Navigating the metabolic maze: anomalies in fatty acid and cholesterol processes in Alzheimer's astrocytes. Alzheimers Res Ther 2024; 16:63. [PMID: 38521950 PMCID: PMC10960454 DOI: 10.1186/s13195-024-01430-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] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, and its underlying mechanisms have been a subject of great interest. The mainstream theory of AD pathology suggests that the disease is primarily associated with tau protein and amyloid-beta (Aβ). However, an increasing body of research has revealed that abnormalities in lipid metabolism may be an important event throughout the pathophysiology of AD. Astrocytes, as important members of the lipid metabolism network in the brain, play a significant role in this event. The study of abnormal lipid metabolism in astrocytes provides a new perspective for understanding the pathogenesis of AD. This review focuses on the abnormal metabolism of fatty acids (FAs) and cholesterol in astrocytes in AD, and discusses it from three perspectives: lipid uptake, intracellular breakdown or synthesis metabolism, and efflux transport. We found that, despite the accumulation of their own fatty acids, astrocytes cannot efficiently uptake fatty acids from neurons, leading to fatty acid accumulation within neurons and resulting in lipotoxicity. In terms of cholesterol metabolism, astrocytes exhibit a decrease in endogenous synthesis due to the accumulation of exogenous cholesterol. Through a thorough investigation of these metabolic abnormalities, we can provide new insights for future therapeutic strategies by literature review to navigate this complex metabolic maze and bring hope to patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chuanying Chen
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Yi Liu
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
39
|
Wang J, Zheng G, Wang L, Meng L, Ren J, Shang L, Li D, Bao Y. Dysregulation of sphingolipid metabolism in pain. Front Pharmacol 2024; 15:1337150. [PMID: 38523645 PMCID: PMC10957601 DOI: 10.3389/fphar.2024.1337150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 02/27/2024] [Indexed: 03/26/2024] Open
Abstract
Pain is a clinical condition that is currently of great concern and is often caused by tissue or nerve damage or occurs as a concomitant symptom of a variety of diseases such as cancer. Severe pain seriously affects the functional status of the body. However, existing pain management programs are not fully satisfactory. Therefore, there is a need to delve deeper into the pathological mechanisms underlying pain generation and to find new targets for drug therapy. Sphingolipids (SLs), as a major component of the bilayer structure of eukaryotic cell membranes, also have powerful signal transduction functions. Sphingolipids are abundant, and their intracellular metabolism constitutes a huge network. Sphingolipids and their various metabolites play significant roles in cell proliferation, differentiation, apoptosis, etc., and have powerful biological activities. The molecules related to sphingolipid metabolism, mainly the core molecule ceramide and the downstream metabolism molecule sphingosine-1-phosphate (S1P), are involved in the specific mechanisms of neurological disorders as well as the onset and progression of various types of pain, and are closely related to a variety of pain-related diseases. Therefore, sphingolipid metabolism can be the focus of research on pain regulation and provide new drug targets and ideas for pain.
Collapse
Affiliation(s)
- Jianfeng Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guangda Zheng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Linfeng Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Linghan Meng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Juanxia Ren
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, China
| | - Lu Shang
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, China
| | - Dongtao Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | | |
Collapse
|
40
|
Liu X, Jin Y, Cheng X, Song Q, Wang Y, He L, Chen T. The relevance between abnormally elevated serum ceramide and cognitive impairment in Alzheimer's disease model mice and its mechanism. Psychopharmacology (Berl) 2024; 241:525-542. [PMID: 38277004 DOI: 10.1007/s00213-024-06530-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
RATIONALE The plasma ceramide levels in Alzheimer's disease (AD) patients are found abnormally elevated, which is related to cognitive decline. OBJECTIVES This research was aimed to investigate the mechanisms of aberrant elevated ceramides in the pathogenesis of AD. RESULTS The ICR mice intracerebroventricularly injected with Aβ1-42 and APP/PS1 transgenic mice were employed as AD mice. The cognitive deficiency, impaired episodic and spatial memory were observed without altered spontaneous ability. The serum levels of p-tau and ceramide were evidently elevated. The modified expressions and activities of glycogen synthase kinase-3β (GSK-3β) and protein phosphatase 2A (PP2A) influenced the serum content of p-tau. The levels of ceramide synthesis-related genes including sptlc1, sptlc2, cers2, and cers6 in the liver of AD mice were increased, while the ceramide degradation-related gene asah2 did not significantly change. The regulations of these genes were conducted by activated nuclear factor kappa-B (NF-κB) signaling. NF-κB, promoted by free fatty acid (FFA), also increased the hepatic concentrations of proinflammatory cytokines. The FFA amount was modulated by fatty acid synthesis-related genes acc1 and srebp-1c. Besides, the decreased levels of pre-proopiomelanocortin (pomc) mRNA and increased agouti-related protein (agrp) mRNA were found in the hypothalamus without significant alteration of melanocortin receptor 4 (MC4R) mRNA. The bioinformatic analyses proved the results using GEO datasets and AlzData. CONCLUSIONS Ceramide was positively related to the increased p-tau and impaired cognitive function. The increased generation of ceramide and endoplasmic reticulum stress in the hypothalamus was positively related to fatty acid synthesis and NF-κB signaling via brain-liver axis.
Collapse
Affiliation(s)
- Xin Liu
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yongzeng Jin
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Xinyi Cheng
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Qinghua Song
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Yanan Wang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.
| | - Tong Chen
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
41
|
Wang X, Li H, Sheng Y, He B, Liu Z, Li W, Yu S, Wang J, Zhang Y, Chen J, Qin L, Meng X. The function of sphingolipids in different pathogenesis of Alzheimer's disease: A comprehensive review. Biomed Pharmacother 2024; 171:116071. [PMID: 38183741 DOI: 10.1016/j.biopha.2023.116071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Sphingolipids (SPLs) represent a highly diverse and structurally complex lipid class. The discussion of SPL metabolism-related issues is of importance in understanding the neuropathological progression of Alzheimer's disease (AD). AD is characterized by the accumulation of extracellular deposits of the amyloid β-peptide (Aβ) and intraneuronal aggregates of the microtubule-associated protein tau. Critical roles of Aβ oligomer deposited and ganglioside GM1 could be formed as "seed" from insoluble GAβ polymer in initiating the pathogenic process, while tau might also mediate SPLs and their toxicity. The interaction between ceramide and α-Synuclein (α-Syn) accelerates the aggregation of ferroptosis and exacerbates the pathogenesis of AD. For instance, reducing the levels of SPLs can mitigate α-Syn accumulation and inhibit AD progression. Meanwhile, loss of SPLs may inhibit the expression of APOE4 and confer protection against AD, while the loss of APOE4 expression also disrupts SPLs homeostasis. Moreover, the heightened activation of sphingomyelinase promotes the ferroptosis signaling pathway, leading to exacerbated AD symptoms. Ferroptosis plays a vital role in the pathological progression of AD by influencing Aβ, tau, APOE, and α-Syn. Conversely, the development of AD also exacerbates the manifestation of ferroptosis and SPLs. We are compiling the emerging techniques (Derivatization and IM-MS) of sphingolipidomics, to overcome the challenges of AD diagnosis and treatment. In this review, we examined the intricate neuro-mechanistic interactions between SPLs and Aβ, tau, α-Syn, APOE, and ferroptosis, mediating the onset of AD. Furthermore, our findings highlight the potential of targeting SPLs as underexplored avenue for devising innovative therapeutic strategies against AD.
Collapse
Affiliation(s)
- Xinyi Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Huaqiang Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Yunjie Sheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Bingqian He
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Zeying Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Wanli Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Shujie Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Jiajing Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Yixin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Jianyu Chen
- Fujian University of Traditional Chinese Medicine, School of Pharmacy, Fuzhou, Fujian 350122, PR China.
| | - Luping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China.
| | - Xiongyu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China.
| |
Collapse
|
42
|
Cui X, Li J, Wang C, Ishaq HM, Zhang R, Yang F. Relationship between sphingolipids-mediated neuroinflammation and alcohol use disorder. Pharmacol Biochem Behav 2024; 235:173695. [PMID: 38128765 DOI: 10.1016/j.pbb.2023.173695] [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: 09/17/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Alcohol use disorder is a chronic recurrent encephalopathy, and its pathogenesis has not been fully understood. Among possible explanations, neuroinflammation caused by the disorders of brain central immune signaling has been identified as one possible mechanism of alcohol use disorder. As the basic components of cells and important bioactive molecules, sphingolipids are essential in regulating many cellular activities. Recent studies have shown that sphingolipids-mediated neuroinflammation may be involved in the development of alcohol use disorder. METHODS PubMed databases were searched for literature on sphingolipids and alcohol use disorder (alcohol abuse, alcohol addiction, alcohol dependence, and alcohol misuse) including evidence of the relationship between sphingolipids-mediated neuroinflammation and alcohol use disorder (formation, withdrawal, treatment). RESULTS Disorders of sphingolipid metabolism, including the different types of sphingolipids and regulatory enzyme activity, have been found in patients with alcohol use disorder as well as animal models, which in turn cause neuro-inflammation in the central nervous system. Thus, these disorders may also be an important mechanism in the development of alcohol use disorder in patients. In addition, different sphingolipids may have different or even reverse effects on alcohol use disorder. CONCLUSIONS The sphingolipids-mediated neuroinflammation plays an important role in the development of alcohol use disorder. This review proposes a potential approach to prevent and treat alcohol use disorders by manipulating sphingolipid metabolism.
Collapse
Affiliation(s)
- XiaoJian Cui
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China; Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - JiaZhen Li
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - ChuanSheng Wang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Hafiz Muhammad Ishaq
- Faculty of Veterinary and Animal Sciences, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - RuiLin Zhang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China.
| | - Fan Yang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China; Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
43
|
Zhao T, Ding T, Sun Z, Shao X, Li S, Lu H, Yuan JH, Guo Z. SPHK1/S1P/S1PR pathway promotes the progression of peritoneal fibrosis by mesothelial-mesenchymal transition. FASEB J 2024; 38:e23417. [PMID: 38226856 DOI: 10.1096/fj.202301323r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/23/2023] [Accepted: 12/27/2023] [Indexed: 01/17/2024]
Abstract
Long-term exposure to non-physiologically compatible dialysate inevitably leads to peritoneal fibrosis (PF) in patients undergoing peritoneal dialysis (PD), and there is no effective prevention or treatment for PF. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid produced after catalysis by sphingosine kinase (SPHK) 1/2 and activates signals through the S1P receptor (S1PR) via autocrine or paracrine. However, the role of SPHK1/S1P/S1PR signaling has never been elucidated in PF. In our research, we investigated S1P levels in peritoneal effluents and demonstrated the role of SPHK1/S1P/S1PR pathway in peritoneal fibrosis. It was found that S1P levels in peritoneal effluents were positively correlated with D/P Cr (r = 0.724, p < .001) and negatively correlated with 4 h ultrafiltration volume (r = -0.457, p < .001). S1PR1 and S1PR3 on peritoneal cells were increased after high glucose exposure in vivo and in vitro. Fingolimod was applied to suppress S1P/S1PR pathway. Fingolimod restored mouse peritoneal function by reducing interstitial hyperplasia, maintaining ultrafiltration volume, reducing peritoneal transport solute rate, and mitigating the protein expression changes of fibronectin, vimentin, α-SMA, and E-cadherin induced by PD and S1P. Fingolimod preserved the morphology of the human peritoneal mesothelial cells, MeT-5A, and moderated the mesothelial-mesenchymal transition (MMT) process. We further delineated that SPHK1 was elevated in peritoneal cells after high glucose exposure and suppression of SPHK1 in MeT-5A cells reduced S1P release. Overexpression of SPHK1 in MeT-5A cells increased S1P levels in the supernatant and fostered the MMT process. PF-543 treatment, targeting SPHK1, alleviated deterioration of mouse peritoneal function. In conclusion, S1P levels in peritoneal effluent were correlated with the deterioration of peritoneal function. SPHK1/S1P/S1PR pathway played an important role in PF.
Collapse
Affiliation(s)
- Tingting Zhao
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, China
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Tao Ding
- Department of Endocrinology, Xizang Military General Hospital, Lhasa City, China
| | - Zhengyu Sun
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, China
| | - Xin Shao
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuangxi Li
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, China
| | - Hongtao Lu
- Department of Nutrition, Naval Medical University, Shanghai, China
| | - Ji-Hang Yuan
- Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Zhiyong Guo
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, China
| |
Collapse
|
44
|
Sood A, Fernandes V, Preeti K, Rajan S, Khatri DK, Singh SB. S1PR2 inhibition mitigates cognitive deficit in diabetic mice by modulating microglial activation via Akt-p53-TIGAR pathway. Int Immunopharmacol 2024; 126:111278. [PMID: 38011768 DOI: 10.1016/j.intimp.2023.111278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
Cognitive deficit is one of the challenging complications of type 2 diabetes. Sphingosine 1- phosphate receptors (S1PRs) have been implicated in various neurodegenerative and metabolic disorders. The association of S1PRs and cognition in type 2 diabetes remains elusive. Microglia-mediated neuronal damage could be the thread propagating cognitive deficit. The effects of S1PR2 inhibition on cognition in high-fat diet and streptozotocin-induced diabetic mice were examined in this work. We further assessed microglial activation and putative microglial polarisation routes. Cognitive function loss was observed after four months of diabetes induction in Type 2 diabetes animal model. JTE013, an S1PR2 inhibitor, was used to assess neuroprotection against cognitive decline and neuroinflammation in vitro and in vivo diabetes model. JTE013 (10 mg/kg) improved synaptic plasticity by upregulating psd95 and synaptophysin while reducing cognitive decline and neuroinflammation. It further enhanced anti-inflammatory microglia in the hippocampus and prefrontal cortex (PFC), as evidenced by increased Arg-1, CD206, and YM-1 levels and decreased iNOS, CD16, and MHCII levels. TIGAR, TP53-induced glycolysis and apoptosis regulator, might facilitate the anti-inflammatory microglial phenotype by promoting oxidative phosphorylation and decreasing apoptosis. However, since p53 is a TIGAR suppressor, inhibiting p53 could be beneficial. S1PR2 inhibition increased p-Akt and TIGAR levels and reduced the levels of p53 in the PFC and hippocampus of type 2 diabetic mice, thereby decreasing apoptosis. In vitro, palmitate was used to imitate sphingolipid dysregulation in BV2 cells, followed by conditioned media exposure to Neuro2A cells. JTE013 rescued the palmitate-induced neuronal apoptosis by promoting the anti-inflammatory microglia. In the present study, we demonstrate that the inhibition of S1PR2 improves cognitive function and skews microglia toward anti-inflammatory phenotype in type 2 diabetic mice, thereby promising to be a potential therapy for neuroinflammation.
Collapse
Affiliation(s)
- Anika Sood
- Department of Pharmacology and Toxicology, NIPER Hyderabad, Hyderabad, Telangana 500037, India
| | - Valencia Fernandes
- Department of Pharmacology and Toxicology, NIPER Hyderabad, Hyderabad, Telangana 500037, India
| | - Kumari Preeti
- Department of Pharmacology and Toxicology, NIPER Hyderabad, Hyderabad, Telangana 500037, India
| | - Shruti Rajan
- Department of Pharmacology and Toxicology, NIPER Hyderabad, Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, NIPER Hyderabad, Hyderabad, Telangana 500037, India.
| | - Shashi Bala Singh
- Department of Pharmacology and Toxicology, NIPER Hyderabad, Hyderabad, Telangana 500037, India.
| |
Collapse
|
45
|
Ma Y, Zhang X, Xuan B, Li D, Yin N, Ning L, Zhou YL, Yan Y, Tong T, Zhu X, Huang X, Hu M, Wang Z, Cui Z, Li H, Wang J, Fang JY, Liu R, Chen H, Hong J. Disruption of CerS6-mediated sphingolipid metabolism by FTO deficiency aggravates ulcerative colitis. Gut 2024; 73:268-281. [PMID: 37734910 DOI: 10.1136/gutjnl-2023-330009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND AND AIMS Deregulation of RNA N6-methyladenosine (m6A) modification in intestinal epithelial cells (IECs) influences intestinal immune cells and leads to intestinal inflammation. We studied the function of fat mass-and obesity-associated protein (FTO), one of the m6A demethylases, in patients with ulcerative colitis (UC). METHODS We analysed colon tissues of Ftoflox/flox; Villin-cre mice and their Ftoflox/flox littermates with dextran sulfate sodium (DSS) using real-time PCR and 16s rRNA sequencing. RNA and methylated RNA immunoprecipitation sequencing were used to analyse immunocytes and IECs. Macrophages were treated with conditioned medium of FTO-knockdown MODE-K cells or sphingosine-1-phosphate (S1P) and analysed for gene expression. Liquid chromatograph mass spectrometry identified C16-ceramide. RESULTS FTO downregulation was identified in our in-house cohort and external cohorts of UC patients. Dysbiosis of gut microbiota, increased infiltration of proinflammatory macrophages, and enhanced differentiation of Th17 cells were observed in Ftoflox/flox;Villin-cre mice under DSS treatment. FTO deficiency resulted in an increase in m6A modification and a decrease in mRNA stability of CerS6, the gene encoding ceramide synthetase, leading to the downregulation of CerS6 and the accumulation of S1P in IECs. Subsequentially, the secretion of S1P by IECs triggered proinflammatory macrophages to secrete serum amyloid A protein 1/3, ultimately inducing Th17 cell differentiation. In addition, through bioinformatic analysis and experimental validation, we identified UC patients with lower FTO expression might respond better to vedolizumab treatment. CONCLUSIONS FTO downregulation promoted UC by decreasing CerS6 expression, leading to increased S1P accumulation in IECs and aggravating colitis via m6A-dependent mechanisms. Lower FTO expression in UC patients may enhance their response to vedolizumab treatment.
Collapse
Affiliation(s)
- Yanru Ma
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyu Zhang
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baoqin Xuan
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Danjie Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, People's Republic of China
| | - Nan Yin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, People's Republic of China
| | - Lijun Ning
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Lu Zhou
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuqing Yan
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianying Tong
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqiang Zhu
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaowen Huang
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Muni Hu
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenhua Wang
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhe Cui
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Huabin Li
- Shanghai Institute of Immunology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, People's Republic of China
| | - Jing-Yuan Fang
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, People's Republic of China
| | - Haoyan Chen
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Hong
- State Key Laboratory of Systems Medicine for Cancer; Key Laboratory of Gastroenterology & Hepatology, Ministry of Health; Division of Gastroenterology and Hepatology; Shanghai Cancer Institute; Shanghai Institute of Digestive Disease; Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
46
|
Chen S, Liang J, Chen D, Huang Q, Sun K, Zhong Y, Lin B, Kong J, Sun J, Gong C, Wang J, Gao Y, Zhang Q, Sun H. Cerebrospinal fluid metabolomic and proteomic characterization of neurologic post-acute sequelae of SARS-CoV-2 infection. Brain Behav Immun 2024; 115:209-222. [PMID: 37858739 DOI: 10.1016/j.bbi.2023.10.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/08/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023] Open
Abstract
The mechanism by which SARS-CoV-2 causes neurological post-acute sequelae of SARS-CoV-2 (neuro-PASC) remains unclear. Herein, we conducted proteomic and metabolomic analyses of cerebrospinal fluid (CSF) samples from 21 neuro-PASC patients, 45 healthy volunteers, and 26 inflammatory neurological diseases patients. Our data showed 69 differentially expressed metabolites and six differentially expressed proteins between neuro-PASC patients and healthy individuals. Elevated sphinganine and ST1A1, sphingolipid metabolism disorder, and attenuated inflammatory responses may contribute to the occurrence of neuro-PASC, whereas decreased levels of 7,8-dihydropterin and activation of steroid hormone biosynthesis may play a role in the repair process. Additionally, a biomarker cohort consisting of sphinganine, 7,8-dihydroneopterin, and ST1A1 was preliminarily demonstrated to have high value in diagnosing neuro-PASC. In summary, our study represents the first attempt to integrate the diagnostic benefits of CSF with the methodological advantages of multi-omics, thereby offering valuable insights into the pathogenesis of neuro-PASC and facilitating the work of neuroscientists in disclosing different neurological dimensions associated with COVID-19.
Collapse
Affiliation(s)
- Shilan Chen
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jianhao Liang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Dingqiang Chen
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Qiyuan Huang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Kaijian Sun
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuxia Zhong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Baojia Lin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jingjing Kong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jiaduo Sun
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Chengfang Gong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jun Wang
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ya Gao
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Qingguo Zhang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
47
|
DiSabato DJ, Marion CM, Mifflin KA, Alfredo AN, Rodgers KA, Kigerl KA, Popovich PG, McTigue DM. System failure: Systemic inflammation following spinal cord injury. Eur J Immunol 2024; 54:e2250274. [PMID: 37822141 PMCID: PMC10919103 DOI: 10.1002/eji.202250274] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/13/2023]
Abstract
Spinal cord injury (SCI) affects hundreds of thousands of people in the United States, and while some effects of the injury are broadly recognized (deficits to locomotion, fine motor control, and quality of life), the systemic consequences of SCI are less well-known. The spinal cord regulates systemic immunological and visceral functions; this control is often disrupted by the injury, resulting in viscera including the gut, spleen, liver, bone marrow, and kidneys experiencing local tissue inflammation and physiological dysfunction. The extent of pathology depends on the injury level, severity, and time post-injury. In this review, we describe immunological and metabolic consequences of SCI across several organs. Since infection and metabolic disorders are primary reasons for reduced lifespan after SCI, it is imperative that research continues to focus on these deleterious aspects of SCI to improve life span and quality of life for individuals with SCI.
Collapse
Affiliation(s)
- Damon J. DiSabato
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
| | - Christina M. Marion
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
| | - Katherine A. Mifflin
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
| | - Anthony N. Alfredo
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Kyleigh A. Rodgers
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Kristina A. Kigerl
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
| | - Phillip G. Popovich
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
| | - Dana M. McTigue
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Belford Center for Spinal Cord Injury, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
48
|
Usenko T, Bezrukova A, Basharova K, Baydakova G, Shagimardanova E, Blatt N, Rizvanov A, Limankin O, Novitskiy M, Shnayder N, Izyumchenko A, Nikolaev M, Zabotina A, Lavrinova A, Kulabukhova D, Nasyrova R, Palchikova E, Zalutskaya N, Miliukhina I, Barbitoff Y, Glotov O, Glotov A, Taraskina A, Neznanov N, Zakharova E, Pchelina S. Altered Sphingolipid Hydrolase Activities and Alpha-Synuclein Level in Late-Onset Schizophrenia. Metabolites 2023; 14:30. [PMID: 38248833 PMCID: PMC10819534 DOI: 10.3390/metabo14010030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Recent data described that patients with lysosomal storage disorders (LSDs) may have clinical schizophrenia (SCZ) features. Disruption of lipid metabolism in SCZ pathogenesis was found. Clinical features of schizophrenia (SCZ) have been demonstrated in patients with several lysosomal storage disorders (LSDs). Taking into account the critical role of lysosomal function for neuronal cells' lysosomal dysfunction could be proposed in SCZ pathogenesis. The current study analyzed lysosomal enzyme activities and the alpha-synuclein level in the blood of patients with late-onset SCZ. In total, 52 SCZ patients with late-onset SCZ, 180 sporadic Parkinson's disease (sPD) patients, and 176 controls were recruited. The enzymatic activity of enzymes associated with mucopolysaccharidosis (alpha-L-Iduronidase (IDUA)), glycogenosis (acid alpha-glucosidase (GAA)) and sphingolipidosis (galactosylceramidase (GALC), glucocerebrosidase (GCase), alpha-galactosidase (GLA), acid sphingomyelinase (ASMase)) and concentration of lysosphingolipids (hexosylsphingosine (HexSph), globotriaosylsphingosine (LysoGb3), and lysosphingomyelin (LysoSM)) were measured using LC-MS/MS. The alpha-synuclein level was estimated in magnetically separated CD45+ blood cells using the enzyme-linked immunosorbent assay (ELISA). Additionally, NGS analysis of 11 LSDs genes was conducted in 21 early-onset SCZ patients and 23 controls using the gene panel PGRNseq-NDD. Decreased ASMase, increased GLA activities, and increased HexSpn, LysoGb3, and LysoSM concentrations along with an accumulation of the alpha-synuclein level were observed in late-onset SCZ patients in comparison to the controls (p < 0.05). Four rare deleterious variants among LSDs genes causing mucopolysaccharidosis type I (IDUA (rs532731688, rs74385837) and type III (HGSNAT (rs766835582)) and sphingolipidosis (metachromatic leukodystrophy (ARSA (rs201251634)) were identified in five patients from the group of early-onset SCZ patients but not in the controls. Our findings supported the role of sphingolipid metabolism in SCZ pathogenesis. Aberrant enzyme activities and compounds of sphingolipids associated with ceramide metabolism may lead to accumulation of alpha-synuclein and may be critical in SCZ pathogenesis.
Collapse
Affiliation(s)
- Tatiana Usenko
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Anastasia Bezrukova
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Katerina Basharova
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Galina Baydakova
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
- Research Center for Medical Genetics, 115478 Moscow, Russia
| | - Elena Shagimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.S.); (N.B.); (A.R.)
| | - Nataliya Blatt
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.S.); (N.B.); (A.R.)
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (E.S.); (N.B.); (A.R.)
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| | - Oleg Limankin
- Psychiatric Hospital No. 1 Named after P. P. Kashchenko, 195009 Saint Petersburg, Russia;
- North-Western Medical University Named after P. I.I. Mechnikov of the Ministry of Health of the Russian Federation, 191015 Saint Petersburg, Russia
| | - Maxim Novitskiy
- Center for Personalized Psychiatry and Neurology of the N.N. V.M. Bekhtereva, 192019 Saint Petersburg, Russia; (M.N.); (N.S.); (R.N.); (N.N.)
| | - Natalia Shnayder
- Center for Personalized Psychiatry and Neurology of the N.N. V.M. Bekhtereva, 192019 Saint Petersburg, Russia; (M.N.); (N.S.); (R.N.); (N.N.)
| | - Artem Izyumchenko
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Mikhail Nikolaev
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Anna Zabotina
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Anna Lavrinova
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Darya Kulabukhova
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Regina Nasyrova
- Center for Personalized Psychiatry and Neurology of the N.N. V.M. Bekhtereva, 192019 Saint Petersburg, Russia; (M.N.); (N.S.); (R.N.); (N.N.)
| | - Ekaterina Palchikova
- V.M. Bekhterev National Medical Research Center Psychiatry and Neurology, 192019 Saint Petersburg, Russia; (E.P.); (N.Z.)
| | - Natalia Zalutskaya
- V.M. Bekhterev National Medical Research Center Psychiatry and Neurology, 192019 Saint Petersburg, Russia; (E.P.); (N.Z.)
| | - Irina Miliukhina
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
- Institute of the Human Brain of RAS, 197022 Saint Petersburg, Russia
| | - Yury Barbitoff
- D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproductology, 199034 Saint Petersburg, Russia; (Y.B.); (O.G.); (A.G.)
- Cerbalab Ltd., 197136 Saint Petersburg, Russia
- Bioinformatics Institute, 197342 Saint Petersburg, Russia
| | - Oleg Glotov
- D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproductology, 199034 Saint Petersburg, Russia; (Y.B.); (O.G.); (A.G.)
- Cerbalab Ltd., 197136 Saint Petersburg, Russia
- Pediatric Research and Clinical Center of Infectious Diseases, 197022 Saint Petersburg, Russia
| | - Andrey Glotov
- D.O. Ott Research Institute for Obstetrics, Gynecology, and Reproductology, 199034 Saint Petersburg, Russia; (Y.B.); (O.G.); (A.G.)
- School of Medicine, St. Petersburg State University, 199034 Saint Petersburg, Russia
| | - Anastasia Taraskina
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| | - Nikolai Neznanov
- Center for Personalized Psychiatry and Neurology of the N.N. V.M. Bekhtereva, 192019 Saint Petersburg, Russia; (M.N.); (N.S.); (R.N.); (N.N.)
- V.M. Bekhterev National Medical Research Center Psychiatry and Neurology, 192019 Saint Petersburg, Russia; (E.P.); (N.Z.)
| | | | - Sofya Pchelina
- Department of Molecular Genetic and Nanobiological Technologies Research Center, Pavlov First Saint-Petersburg State Medical University, 197022 Saint Petersburg, Russia; (T.U.); (A.B.); (A.I.); (M.N.); (A.Z.); (D.K.); (I.M.); (A.T.); (S.P.)
- Petersburg Nuclear Physics Institute Named by B.P. Konstantinov of National Research Centre Kurchatov Institute, 188300 Gatchina, Russia (G.B.); (A.L.)
| |
Collapse
|
49
|
Wang W, Zhao Y, Zhu G. The role of sphingosine-1-phosphate in the development and progression of Parkinson's disease. Front Cell Neurosci 2023; 17:1288437. [PMID: 38179204 PMCID: PMC10764561 DOI: 10.3389/fncel.2023.1288437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Parkinson's disease (PD) could be viewed as a proteinopathy caused by changes in lipids, whereby modifications in lipid metabolism may lead to protein alterations, such as the accumulation of alpha-synuclein (α-syn), ultimately resulting in neurodegeneration. Although the loss of dopaminergic neurons in the substantia nigra is the major clinical manifestation of PD, the etiology of it is largely unknown. Increasing evidence has highlighted the important role of lipids in the pathophysiology of PD. Sphingosine-1-phosphate (S1P), a signaling lipid, has been suggested to have a potential association with the advancement and worsening of PD. Therefore, better understanding the mechanisms and regulatory proteins is of high interest. Most interestingly, S1P appears to be an important target to offers a new strategy for the diagnosis and treatment of PD. In this review, we first introduce the basic situation of S1P structure, function and regulation, with a special focus on the several pathways. We then briefly describe the regulation of S1P signaling pathway on cells and make a special focused on the cell growth, proliferation and apoptosis, etc. Finally, we discuss the function of S1P as potential therapeutic target to improve the clinical symptoms of PD, and even prevent the progression of the PD. In the context of PD, the functions of S1P modulators have been extensively elucidated. In conclusion, S1P modulators represent a novel and promising therapeutic principle and therapeutic method for PD. However, more research is required before these drugs can be considered as a standard treatment option for PD.
Collapse
Affiliation(s)
- Wang Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guoxue Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
50
|
Yang H, Shen M, Zhang Q, Li Y, Tan X, Li X, Chen H, Wu L, He S, Zhu X. Transcriptome and metabolomics analysis of adaptive mechanism of Chinese mitten crab (Eriocheir sinensis) to aflatoxin B1. PLoS One 2023; 18:e0295291. [PMID: 38060597 PMCID: PMC10703319 DOI: 10.1371/journal.pone.0295291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/19/2023] [Indexed: 12/18/2023] Open
Abstract
Aflatoxin B1 (AFB1), with the strong toxicity and carcinogenicity, has been reported to great toxicity to the liver and other organs of animals. It cause huge economic losses to breeding industry, including the aquaculture industry. Chinese mitten crabs (Eriocheir sinensis), as one of important species of freshwater aquaculture in China, are deeply disturbed by it. However, the molecular and metabolic mechanisms of hepatopancreas and ovary in crabs underlying coping ability are still unclear. Hence, we conducted targeted injection experiment with or without AFB1, and comprehensively analyzed transcriptome and metabolomics of hepatopancreas and ovary. As a result, 210 and 250 DEGs were identified in the L-C vs. L-30 m and L-C vs. L-60 m comparison, among which 14 common DEGs were related to six major functional categories, including antibacterial and detoxification, ATP energy reaction, redox reaction, nerve reaction, liver injury repair and immune reaction. A total of 228 and 401 DAMs in the ML-C vs. ML-30 m and ML-C vs. ML-60 m comparison both enriched 12 pathways, with clear functions of cutin, suberine and wax biosynthesis, tyrosine metabolism, purine metabolism, nucleotide metabolism, glycine, serine and threonine metabolism, ABC transporters and tryptophan metabolism. Integrated analysis of metabolomics and transcriptome in hepatopancreas discovered three Co-enriched pathways, including steroid biosynthesis, glycine, serine and threonine metabolism, and sphingolipid metabolism. In summary, the expression levels and functions of related genes and metabolites reveal the regulatory mechanism of Chinese mitten crab (Eriocheir sinensis) adaptability to the Aflatoxin B1, and the findings contribute to a new perspective for understanding Aflatoxin B1 and provide some ideas for dealing with it.
Collapse
Affiliation(s)
- Hongsheng Yang
- Fishery Analysis and Testing Center of Jiangsu Province, Nanjing, Jiangsu, China
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, Jiangsu, China
| | - Meifang Shen
- Fishery Analysis and Testing Center of Jiangsu Province, Nanjing, Jiangsu, China
| | - Qiuyun Zhang
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, Jiangsu, China
| | - Yifeng Li
- College of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Xiuhui Tan
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, Jiangsu, China
| | - Xuguang Li
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, Jiangsu, China
| | - Huimin Chen
- SCIEX Analytical Instrument Trading Co., Shanghai, China
| | - Lei Wu
- Yitian Technologies Corporation, Nanjing, Jiangsu, China
| | - Shaofang He
- Yitian Technologies Corporation, Nanjing, Jiangsu, China
| | - Xiaohua Zhu
- Fishery Analysis and Testing Center of Jiangsu Province, Nanjing, Jiangsu, China
| |
Collapse
|