1
|
Lee H, Nakahashi-Oda C, Lyu W, Tanaka M, Rai A, Muramoto Y, Wang Y, Mizuno S, Shibuya K, Shibuya A. Inhibitory immunoreceptors CD300a and CD300lf cooperate to regulate mast cell activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae030. [PMID: 40073110 DOI: 10.1093/jimmun/vkae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/07/2024] [Indexed: 03/14/2025]
Abstract
Mast cells (MCs) play a central role in allergic immune responses. MC activation is regulated by several inhibitory immunoreceptors. The CD300 family members CD300a and CD300lf recognize phospholipid ligands and inhibit the FcεRI-mediated activating signal in MCs. While CD300a binds to phosphatidylserine (PS) to inhibit MCs activation, CD300lf function is less clear due to its ability to bind with ceramide and PS. Moreover, it also remains blurring whether CD300a and CD300lf function independently, cooperatively, or by interfering with each other in regulating MC activation. Using imaging and flow cytometric analyses of bone marrow-derived cultured MCs (BMMCs) from wild-type (WT), Cd300a-/-, Cd300lf-/-, and Cd300a-/-Cd300lf-/- mice, we show that CD300lf and CD300a colocalized with PS externalized to the outer leaflet of the plasma membrane with a polar formation upon activation, and CD300lf cooperates with CD300a to inhibit BMMCs activation. CD300lf also colocalized with extracellular ceramide in addition to the internal PS on the cell surface, which results in stronger inhibition of MC activation than CD300lf binding to PS alone. Similarly, although both Cd300a-/- and Cd300lf-/- mice showed decreased rectal temperatures compared with WT mice in the model of passive systemic anaphylaxis, Cd300a-/-Cd300lf-/- mice showed lower rectal temperature than either Cd300a-/- or Cd300lf-/- mice. Our results demonstrate the cooperativity of multiple inhibitory receptors expressed on MCs and their regulatory functions upon binding to respective ligands.
Collapse
Affiliation(s)
- Hanbin Lee
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Ph.D. Program in Human Biology, Graduate School of Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Chigusa Nakahashi-Oda
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Wenxin Lyu
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Ph.D. Program in Human Biology, Graduate School of Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Mamoru Tanaka
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Master's Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akiyoshi Rai
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoichi Muramoto
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- School of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yaqiu Wang
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Ph.D. Program in Human Biology, Graduate School of Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuko Shibuya
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Shibuya
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), Tsukuba, Ibaraki, Japan
| |
Collapse
|
2
|
Deng C, Xiao Y, Zhao X, Li H, Chen Y, Ai K, Jiang T, Wei J, Chen X, Lei G, Zeng C. Sequential Targeting Chondroitin Sulfate-Bilirubin Nanomedicine Attenuates Osteoarthritis via Reprogramming Lipid Metabolism in M1 Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411911. [PMID: 39792653 PMCID: PMC11884591 DOI: 10.1002/advs.202411911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/16/2024] [Indexed: 01/12/2025]
Abstract
The infiltration and excessive polarization of M1 macrophages contribute to the induction and persistence of low-grade inflammation in joint-related degenerative diseases such as osteoarthritis (OA). The lipid metabolism dysregulation promotes M1 macrophage polarization by coordinating the compensatory pathways of the inflammatory and oxidative stress responses. Here, a self-assembling, licofelone-loaded nanoparticle (termed LCF-CSBN), comprising chondroitin sulfate and bilirubin joined by an ethylenediamine linker, is developed to selectively reprogram lipid metabolism in macrophage activation. LCF-CSBN is internalized by M1 macrophages via CD44-mediated endocytosis and targets the Golgi apparatus accompanied with the reactive oxygen species-responsive release of licofelone (LCF, dual inhibitor of arachidonic acid metabolism). LCF-CSBN effectively promotes M1 to M2 macrophage transition by reprogramming the Golgi apparatus-related sphingolipid metabolism and arachidonic acid metabolism. Intra-articularly injected LCF-CSBN retains in the joint for up to 28 days and accumulates into M1 macrophages. Moreover, LCF-CSBN can effectively attenuate joint inflammation, oxidative stress, and cartilage degeneration in OA model rats. These findings indicate the promising potential of lipid-metabolism-reprogramming LCF-CSBN in the targeted therapy of OA.
Collapse
Affiliation(s)
- Caifeng Deng
- Department of Orthopaedics, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of Education, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yongbing Xiao
- Department of Orthopaedics, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of Education, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Xuan Zhao
- Department of Orthopaedics, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of Education, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Hui Li
- Department of Orthopaedics, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of Education, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yuxiao Chen
- Department of Orthopaedics, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of Education, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Kelong Ai
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangshaHunan410008China
| | - Ting Jiang
- Department of Orthopaedics, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of Education, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Jie Wei
- Department of Orthopaedics, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of Education, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingapore119074Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of MedicineNational University of SingaporeSingapore117599Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of MedicineNational University of SingaporeSingapore117597Singapore
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of Education, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of Education, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| |
Collapse
|
3
|
Pellegrino R, Imperio G, De Costanzo I, Izzo M, Landa F, Tambaro A, Gravina AG, Federico A. Small Molecules in the Treatment of Acute Severe Ulcerative Colitis: A Review of Current Evidence. Pharmaceuticals (Basel) 2025; 18:308. [PMID: 40143087 PMCID: PMC11944803 DOI: 10.3390/ph18030308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease in which one-quarter of patients are at risk of developing a severe form of the disease known as acute severe UC (ASUC). This condition exposes patients to serious complications, including toxic megacolon, surgical intervention, and even death. The current therapeutic strategy relies on time-dependent, multi-step algorithms that integrate systemic corticosteroids, calcineurin inhibitors, and biologic agents (specifically infliximab) as medical therapy aimed at avoiding colectomy. Despite this approach, a significant proportion of patients fail to respond to either corticosteroids or infliximab and may require alternative therapeutic options if there is no urgent surgical necessity. These alternatives include other biologics or emerging small molecules, although the evidence supporting these treatments remains extremely low, even considering their well-documented and promising efficacy and safety in moderate-to-severe UC. Conversely, it is necessary to investigate whether infliximab can be effectively replaced or surpassed by other approved biological agents and small molecules as first-line therapy after steroid resistance. This review aims to summarise the available evidence on small molecules, specifically Janus kinase inhibitors and sphingosine-1-phosphate receptor modulators.
Collapse
Affiliation(s)
- Raffaele Pellegrino
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. de Crecchio, 80138 Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Ouyang Z, Tan Z, Ali U, Zhang Y, Li B, Yao X, Yang B, Guo L. Ceramide-1-phosphate enhances defense responses against Sclerotinia sclerotiorum in Brassica napus. PLANT PHYSIOLOGY 2025; 197:kiae649. [PMID: 39928582 DOI: 10.1093/plphys/kiae649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 10/27/2024] [Indexed: 02/12/2025]
Abstract
Sclerotinia stem rot caused by Sclerotinia sclerotiorum is one of the most severe diseases affecting the growth and production of Brassica napus. Sphingolipid metabolism plays a crucial role in plant response to pathogens. In this study, we show that ceramide kinase (CERK) is significantly induced during S. sclerotiorum infection to produce higher levels of ceramide-1-phosphate (C1P) in B. napus. The balance between ceramide (Cer) and C1P affects plant resistance to S. sclerotiorum, with CERK mutant lines exhibiting greater susceptibility to S. sclerotiorum and CERK overexpression lines showing enhanced resistance to this pathogen. Moreover, we identified candidate C1P-binding proteins by proteomic analysis and determined that C1P binds to and promotes the activity of a Gly-Asp-Ser-Leu lipase protein involved in B. napus resistance to S. sclerotiorum infection. In conclusion, our results indicate that C1P plays a key role in S. sclerotiorum resistance through metabolic regulation and signal transduction in B. napus.
Collapse
Affiliation(s)
- Zhewen Ouyang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Zengdong Tan
- Yazhouwan National Laboratory, Sanya 572025, China
| | - Usman Ali
- State Key Laboratory of Wheat and Maize Crop Science, Henan Agricultural University, Zhengzhou 450046, China
| | - Ying Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- The Provincial Key Lab of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Bo Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- The Provincial Key Lab of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xuan Yao
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- Yazhouwan National Laboratory, Sanya 572025, China
| | - Bao Yang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- Department of Biology, University of Missouri, St. Louis, MO 63121, USA
- Donald Danforth Plant Science Center, St. Louis, MO 63132, USA
| | - Liang Guo
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- Yazhouwan National Laboratory, Sanya 572025, China
| |
Collapse
|
5
|
Esplin ED, Hanson C, Wu S, Horning AM, Barapour N, Nevins SA, Jiang L, Contrepois K, Lee H, Guha TK, Hu Z, Laquindanum R, Mills MA, Chaib H, Chiu R, Jian R, Chan J, Ellenberger M, Becker WR, Bahmani B, Khan A, Michael B, Weimer AK, Esplin DG, Shen J, Lancaster S, Monte E, Karathanos TV, Ladabaum U, Longacre TA, Kundaje A, Curtis C, Greenleaf WJ, Ford JM, Snyder MP. Multiomic analysis of familial adenomatous polyposis reveals molecular pathways associated with early tumorigenesis. NATURE CANCER 2024; 5:1737-1753. [PMID: 39478120 PMCID: PMC11584401 DOI: 10.1038/s43018-024-00831-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 08/29/2024] [Indexed: 11/24/2024]
Abstract
Familial adenomatous polyposis (FAP) is a genetic disease causing hundreds of premalignant polyps in affected persons and is an ideal model to study transitions of early precancer states to colorectal cancer (CRC). We performed deep multiomic profiling of 93 samples, including normal mucosa, benign polyps and dysplastic polyps, from six persons with FAP. Transcriptomic, proteomic, metabolomic and lipidomic analyses revealed a dynamic choreography of thousands of molecular and cellular events that occur during precancerous transitions toward cancer formation. These involve processes such as cell proliferation, immune response, metabolic alterations (including amino acids and lipids), hormones and extracellular matrix proteins. Interestingly, activation of the arachidonic acid pathway was found to occur early in hyperplasia; this pathway is targeted by aspirin and other nonsteroidal anti-inflammatory drugs, a preventative treatment under investigation in persons with FAP. Overall, our results reveal key genomic, cellular and molecular events during the earliest steps in CRC formation and potential mechanisms of pharmaceutical prophylaxis.
Collapse
Affiliation(s)
- Edward D Esplin
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Casey Hanson
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Si Wu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Aaron M Horning
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Nasim Barapour
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Lihua Jiang
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Hayan Lee
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Tuhin K Guha
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Zheng Hu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | | | - Meredith A Mills
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Hassan Chaib
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Roxanne Chiu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Joanne Chan
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Winston R Becker
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Bahareh Bahmani
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Aziz Khan
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, USA
| | - Basil Michael
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Annika K Weimer
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Jeanne Shen
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Samuel Lancaster
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Emma Monte
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Uri Ladabaum
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Teri A Longacre
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Christina Curtis
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - James M Ford
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA.
- Department of Medicine, Stanford School of Medicine, Stanford, CA, USA.
| | - Michael P Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Chen W, Han Y, Xu Y, Wang T, Wang Y, Chen X, Qiu X, Li W, Li H, Fan Y, Yao Y, Zhu T. Fine particulate matter exposure and systemic inflammation: A potential mediating role of bioactive lipids. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 931:172993. [PMID: 38719056 DOI: 10.1016/j.scitotenv.2024.172993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/20/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
Inflammation is a key mechanism underlying the adverse health effects of exposure to fine particulate matter (PM2.5). Bioactive lipids in the arachidonic acid (ARA) pathway are important in the regulation of inflammation and are reportedly altered by PM2.5 exposure. Ceramide-1-phosphate (C1P), a class of sphingolipids, is required to initiate ARA metabolism. We examined the role of C1P in the alteration of ARA metabolism after PM2.5 exposure and explored whether changes in the ARA pathway promoted systemic inflammation based on a panel study involving 112 older adults in Beijing, China. Ambient PM2.5 levels were continuously monitored at a fixed station from 2013 to 2015. Serum cytokine levels were measured to assess systemic inflammation. Multiple bioactive lipids in the ARA pathway and three subtypes of C1P were quantified in blood samples. Mediation analyses were performed to test the hypotheses. We observed that PM2.5 exposure was positively associated with inflammatory cytokines and the three subtypes of C1P. Mediation analyses showed that C1P significantly mediated the associations of ARA and 5, 6-dihydroxyeicosatrienoic acid (5, 6-DHET), an ARA metabolite, with PM2.5 exposure. ARA, 5, 6-DHET, and leukotriene B4 mediated systemic inflammatory response to PM2.5 exposure. For example, C1P C16:0 (a subtype of C1P) mediated a 12.9 % (95 % confidence interval: 3.7 %, 32.5 %) increase in ARA associated with 3-day moving average PM2.5 exposure, and ARA mediated a 27.1 % (7.8 %, 61.2 %) change in interleukin-8 associated with 7-day moving average PM2.5 exposure. Our study indicates that bioactive lipids in the ARA and sphingolipid metabolic pathways may mediate systemic inflammation after PM2.5 exposure.
Collapse
Affiliation(s)
- Wu Chen
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yiqun Han
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Environmental Research Group, MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Yifan Xu
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Teng Wang
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Yanwen Wang
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xi Chen
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Hebei Technology Innovation Center of Human Settlement in Green Building (TCHS), Shenzhen Institute of Building Research Co., Ltd., Xiongan, Hebei, China
| | - Xinghua Qiu
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Weiju Li
- Peking University Hospital, Peking University, Beijing, China
| | - Haonan Li
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Yunfei Fan
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; China National Environmental Monitoring Centre, Beijing, China
| | - Yuan Yao
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Department of Environmental Health Sciences, Jonathan and Karin Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Tong Zhu
- BIC-ESAT and SKL-ESPC, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China.
| |
Collapse
|
9
|
Wajapeyee N, Beamon TC, Gupta R. Roles and therapeutic targeting of ceramide metabolism in cancer. Mol Metab 2024; 83:101936. [PMID: 38599378 PMCID: PMC11031839 DOI: 10.1016/j.molmet.2024.101936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Ceramides are sphingolipids that act as signaling molecules involved in regulating cellular processes including apoptosis, proliferation, and metabolism. Deregulation of ceramide metabolism contributes to cancer development and progression. Therefore, regulation of ceramide levels in cancer cells is being explored as a new approach for cancer therapy. SCOPE OF THE REVIEW This review discusses the multiple roles of ceramides in cancer cells and strategies to modulate ceramide levels for cancer therapy. Ceramides attenuate cell survival signaling and metabolic pathways, while activating apoptotic mechanisms, making them tumor-suppressive. Approaches to increase ceramide levels in cancer cells include using synthetic analogs, inhibiting ceramide degradation, and activating ceramide synthesis. We also highlight combination therapies such as use of ceramide modulators with chemotherapies, immunotherapies, apoptosis inducers, and anti-angiogenics, which offer synergistic antitumor effects. Additionally, we also describe ongoing clinical trials evaluating ceramide nanoliposomes and analogs. Finally, we discuss the challenges of these therapeutic approaches including the complexity of ceramide metabolism, targeted delivery, cancer heterogeneity, resistance mechanisms, and long-term safety. MAJOR CONCLUSIONS Ceramide-based therapy is a potentially promising approach for cancer therapy. However, overcoming hurdles in pharmacokinetics, specificity, and resistance is needed to optimize its efficacy and safety. This requires comprehensive preclinical/clinical studies into ceramide signaling, formulations, and combination therapies. Ceramide modulation offers opportunities for developing novel cancer treatments, but a deeper understanding of ceramide biology is vital to advance its clinical applications.
Collapse
Affiliation(s)
- Narendra Wajapeyee
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| | - Teresa Chiyanne Beamon
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
10
|
Yang X, Wang K, Shen P, Zhou T, Xu Y, Chen Y, Li Y, Yao Y, Gong Z, Duan R, Jing L, Jia Y. Association of plasma sphingosine-1-phosphate levels with disease severity and prognosis after intracerebral hemorrhage. Front Neurol 2024; 15:1365902. [PMID: 38633536 PMCID: PMC11021779 DOI: 10.3389/fneur.2024.1365902] [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: 01/05/2024] [Accepted: 03/21/2024] [Indexed: 04/19/2024] Open
Abstract
Purpose Sphingosine-1-phosphate (S1P) is a signaling lipid involved in many biological processes, including inflammatory and immune regulatory responses. The study aimed to determine whether admission S1P levels are associated with disease severity and prognosis after spontaneous intracerebral hemorrhage (ICH). Methods Data of 134 patients with spontaneous ICH and 120 healthy controls were obtained from Biological Resource Sample Database of Intracerebral Hemorrhage at the First Affiliated Hospital of Zhengzhou University. Plasma S1P levels were measured. Regression analyses were used to analyze the association between S1P levels and admission and 90-day modified Rankin scale (mRS) scores. Receiver operating characteristic (ROC) curves assessed the predictive value of S1P levels for ICH severity and prognosis. Results Patients with ICH exhibited elevated plasma S1P levels compared to the control group (median 286.95 vs. 239.80 ng/mL, p < 0.001). When divided patients into mild-to-moderate and severe groups according to their mRS scores both at admission and discharge, S1P levels were significantly elevated in the severe group compared to the mild-to-moderate group (admission 259.30 vs. 300.54, p < 0.001; 90-day 275.24 vs. 303.25, p < 0.001). The patients were divided into three groups with different concentration gradients, which showed significant statistical differences in admission mRS scores (3 vs. 4 vs. 5, p < 0.001), 90-day mRS scores (2.5 vs. 3 vs. 4, p < 0.001), consciousness disorders (45.5% vs. 68.2% vs. 69.6%, p = 0.033), ICU admission (29.5% vs. 59.1% vs. 89.1%, p < 0.001), surgery (15.9% vs. 47.7% vs. 82.6%, p < 0.001), intraventricular hemorrhages (27.3% vs. 61.4% vs. 65.2%, p < 0.001) and pulmonary infection (25% vs. 47.7% vs. 84.8%, p < 0.001). Multivariate analysis displayed that S1P level was an independent risk factor for disease severity (OR = 1.037, 95% CI = 1.020-1.054, p < 0.001) and prognosis (OR = 1.018, 95% CI = 1.006-1.030, p = 0.003). ROC curves revealed a predictive value of S1P levels with an area under the curve of 0.7952 (95% CI = 0.7144-0.8759, p < 0.001) for disease severity and 0.7105 (95% CI = 0.6227-0.7983, p < 0.001) for prognosis. Conclusion Higher admission S1P is associated with worse initial disease severity and 90-day functional outcomes in intracerebral hemorrhage.
Collapse
Affiliation(s)
- Xuan Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaixin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ping Shen
- Department of Neurology, Xinzheng Huaxin Minsheng Hospital, Zhengzhou, Henan, China
| | - Tong Zhou
- Department of Neurology, Huaiyang County People’s Hospital, Zhoukou, Henan, China
| | - Yudi Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yufei Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanfei Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaobing Yao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhe Gong
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ranran Duan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijun Jing
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanjie Jia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Seal A, Hughes M, Wei F, Pugazhendhi AS, Ngo C, Ruiz J, Schwartzman JD, Coathup MJ. Sphingolipid-Induced Bone Regulation and Its Emerging Role in Dysfunction Due to Disease and Infection. Int J Mol Sci 2024; 25:3024. [PMID: 38474268 PMCID: PMC10932382 DOI: 10.3390/ijms25053024] [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: 02/09/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
The human skeleton is a metabolically active system that is constantly regenerating via the tightly regulated and highly coordinated processes of bone resorption and formation. Emerging evidence reveals fascinating new insights into the role of sphingolipids, including sphingomyelin, sphingosine, ceramide, and sphingosine-1-phosphate, in bone homeostasis. Sphingolipids are a major class of highly bioactive lipids able to activate distinct protein targets including, lipases, phosphatases, and kinases, thereby conferring distinct cellular functions beyond energy metabolism. Lipids are known to contribute to the progression of chronic inflammation, and notably, an increase in bone marrow adiposity parallel to elevated bone loss is observed in most pathological bone conditions, including aging, rheumatoid arthritis, osteoarthritis, and osteomyelitis. Of the numerous classes of lipids that form, sphingolipids are considered among the most deleterious. This review highlights the important primary role of sphingolipids in bone homeostasis and how dysregulation of these bioactive metabolites appears central to many chronic bone-related diseases. Further, their contribution to the invasion, virulence, and colonization of both viral and bacterial host cell infections is also discussed. Many unmet clinical needs remain, and data to date suggest the future use of sphingolipid-targeted therapy to regulate bone dysfunction due to a variety of diseases or infection are highly promising. However, deciphering the biochemical and molecular mechanisms of this diverse and extremely complex sphingolipidome, both in terms of bone health and disease, is considered the next frontier in the field.
Collapse
Affiliation(s)
- Anouska Seal
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
| | - Megan Hughes
- School of Biosciences, Cardiff University, Cardiff CF10 3AT, UK;
| | - Fei Wei
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | - Abinaya S. Pugazhendhi
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | - Christopher Ngo
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | - Jonathan Ruiz
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| | | | - Melanie J. Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA; (A.S.); (F.W.); (A.S.P.); (C.N.)
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA (J.D.S.)
| |
Collapse
|
12
|
Eggers E, Crouss T, Lipetskaia L, DiSanto M. Urinary Sphingosine-1-Phosphate as a Biomarker for Bladder Pain Syndrome. UROGYNECOLOGY (PHILADELPHIA, PA.) 2024; 30:374-380. [PMID: 38484256 DOI: 10.1097/spv.0000000000001473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
IMPORTANCE Sphingosine-1-phosphate (S1P) is a signaling molecule involved in inflammation and bladder contraction. OBJECTIVES The aims of this case-control pilot study were to compare urinary S1P concentrations in bladder pain syndrome (BPS) participants to controls and determine whether these concentrations correlate with disease severity and duration. STUDY DESIGN Adult females with BPS and controls were enrolled. Bladder pain syndrome participants completed an O'Leary-Sant questionnaire. Information on duration of symptoms and treatment history was obtained. Urinary S1P and creatinine concentrations were determined. Mann-Whitney U tests were used to compare groups, and Spearman correlation was used to test for associations between concentrations and duration and severity of symptoms. RESULTS Twenty-five participants were in each group. Median S1P concentration was 1,225 ng/dL in the BPS group and 2,183 ng/dL in the control group, which was significantly different (P < 0.0001). This difference did not persist when normalized to urinary creatinine (P = 0.58). No differences were noted in urinary S1P concentrations between treated and untreated participants (P = 0.53) or with symptom scores of 13 or greater and less than 13 (P = 0.69). Sphingosine-1-phosphate levels did not correlate with O'Leary-Sant scores (P = 0.08) or duration of symptoms (P = 0.67). Results did not change when using S1P concentrations normalized to creatinine. CONCLUSIONS This study demonstrated successful quantification of human urinary S1P concentrations. A difference in urinary S1P was found between BPS participants and controls but not when normalized to creatinine. While this is the first study to investigate urinary S1P as a biomarker for BPS, results suggest that it may have a potential role as a biomarker requiring further research.
Collapse
Affiliation(s)
- Erica Eggers
- From the Department of Obstetrics and Gynecology, Division of Female Pelvic Medicine and Reconstructive Surgery, Cooper University Healthcare
| | - Tess Crouss
- From the Department of Obstetrics and Gynecology, Division of Female Pelvic Medicine and Reconstructive Surgery, Cooper University Healthcare
| | - Lioudmila Lipetskaia
- From the Department of Obstetrics and Gynecology, Division of Female Pelvic Medicine and Reconstructive Surgery, Cooper University Healthcare
| | - Michael DiSanto
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ
| |
Collapse
|
13
|
Norris AC, Yazlovitskaya EM, Zhu L, Rose BS, May JC, Gibson-Corley KN, McLean JA, Stafford JM, Graham TR. Deficiency of the lipid flippase ATP10A causes diet-induced dyslipidemia in female mice. Sci Rep 2024; 14:343. [PMID: 38172157 PMCID: PMC10764864 DOI: 10.1038/s41598-023-50360-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Genetic association studies have linked ATP10A and closely related type IV P-type ATPases (P4-ATPases) to insulin resistance and vascular complications, such as atherosclerosis. ATP10A translocates phosphatidylcholine and glucosylceramide across cell membranes, and these lipids or their metabolites play important roles in signal transduction pathways regulating metabolism. However, the influence of ATP10A on lipid metabolism in mice has not been explored. Here, we generated gene-specific Atp10A knockout mice and show that Atp10A-/- mice fed a high-fat diet did not gain excess weight relative to wild-type littermates. However, Atp10A-/- mice displayed female-specific dyslipidemia characterized by elevated plasma triglycerides, free fatty acids and cholesterol, as well as altered VLDL and HDL properties. We also observed increased circulating levels of several sphingolipid species along with reduced levels of eicosanoids and bile acids. The Atp10A-/- mice also displayed hepatic insulin resistance without perturbations to whole-body glucose homeostasis. Thus, ATP10A has a sex-specific role in regulating plasma lipid composition and maintaining hepatic liver insulin sensitivity in mice.
Collapse
Affiliation(s)
- Adriana C Norris
- Department of Biological Sciences, Vanderbilt University, 465 21St Ave S, Nashville, TN, 37212, USA
| | - Eugenia M Yazlovitskaya
- Department of Biological Sciences, Vanderbilt University, 465 21St Ave S, Nashville, TN, 37212, USA
| | - Lin Zhu
- Division of Endocrinology, Diabetes and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bailey S Rose
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA
| | - Jody C May
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA
| | - Katherine N Gibson-Corley
- Division of Comparative Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John A McLean
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, USA
| | - John M Stafford
- Division of Endocrinology, Diabetes and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Todd R Graham
- Department of Biological Sciences, Vanderbilt University, 465 21St Ave S, Nashville, TN, 37212, USA.
| |
Collapse
|
14
|
Jamjoum R, Majumder S, Issleny B, Stiban J. Mysterious sphingolipids: metabolic interrelationships at the center of pathophysiology. Front Physiol 2024; 14:1229108. [PMID: 38235387 PMCID: PMC10791800 DOI: 10.3389/fphys.2023.1229108] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/27/2023] [Indexed: 01/19/2024] Open
Abstract
Metabolic pathways are complex and intertwined. Deficiencies in one or more enzymes in a given pathway are directly linked with genetic diseases, most of them having devastating manifestations. The metabolic pathways undertaken by sphingolipids are diverse and elaborate with ceramide species serving as the hubs of sphingolipid intermediary metabolism and function. Sphingolipids are bioactive lipids that serve a multitude of cellular functions. Being pleiotropic in function, deficiency or overproduction of certain sphingolipids is associated with many genetic and chronic diseases. In this up-to-date review article, we strive to gather recent scientific evidence about sphingolipid metabolism, its enzymes, and regulation. We shed light on the importance of sphingolipid metabolism in a variety of genetic diseases and in nervous and immune system ailments. This is a comprehensive review of the state of the field of sphingolipid biochemistry.
Collapse
Affiliation(s)
- Rama Jamjoum
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | - Saurav Majumder
- National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Rockville, MD, United States
| | - Batoul Issleny
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | - Johnny Stiban
- Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine
| |
Collapse
|
15
|
Norris AC, Yazlovitskaya EM, Zhu L, Rose BS, May JC, Gibson-Corley KN, McLean JA, Stafford JM, Graham TR. Deficiency of the lipid flippase ATP10A causes diet-induced dyslipidemia in female mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545392. [PMID: 37398141 PMCID: PMC10312798 DOI: 10.1101/2023.06.16.545392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Genetic association studies have linked ATP10A and closely related type IV P-type ATPases (P4-ATPases) to insulin resistance and vascular complications, such as atherosclerosis. ATP10A translocates phosphatidylcholine and glucosylceramide across cell membranes, and these lipids or their metabolites play important roles in signal transduction pathways regulating metabolism. However, the influence of ATP10A on lipid metabolism in mice has not been explored. Here, we generated gene-specific Atp10A knockout mice and show that Atp10A-/- mice fed a high-fat diet did not gain excess weight relative to wild-type littermates. However, Atp10A-/- mice displayed female-specific dyslipidemia characterized by elevated plasma triglycerides, free fatty acids and cholesterol, as well as altered VLDL and HDL properties. We also observed increased circulating levels of several sphingolipid species along with reduced levels of eicosanoids and bile acids. The Atp10A-/- mice also displayed hepatic insulin resistance without perturbations to whole-body glucose homeostasis. Thus, ATP10A has a sex-specific role in regulating plasma lipid composition and maintaining hepatic liver insulin sensitivity in mice.
Collapse
Affiliation(s)
- Adriana C. Norris
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Lin Zhu
- Division of Endocrinology, Diabetes and Metabolism, Vanderbilt University Medical Center, USA
| | - Bailey S. Rose
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, USA
| | - Jody C. May
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, USA
| | - Katherine N. Gibson-Corley
- Division of Comparative Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - John A. McLean
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, USA
- Center for Innovative Technology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee, USA
| | - John M. Stafford
- Division of Endocrinology, Diabetes and Metabolism, Vanderbilt University Medical Center, USA
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Tennessee, USA
| | - Todd R. Graham
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
16
|
Ya'ar Bar S, Pintel N, Abd Alghne H, Khattib H, Avni D. The therapeutic potential of sphingolipids for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1224743. [PMID: 37608809 PMCID: PMC10440740 DOI: 10.3389/fcvm.2023.1224743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/17/2023] [Indexed: 08/24/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide and Inflammation plays a critical role in the development of CVD. Despite considerable progress in understanding the underlying mechanisms and various treatment options available, significant gaps in therapy necessitate the identification of novel therapeutic targets. Sphingolipids are a family of lipids that have gained attention in recent years as important players in CVDs and the inflammatory processes that underlie their development. As preclinical studies have shown that targeting sphingolipids can modulate inflammation and ameliorate CVDs, targeting sphingolipids has emerged as a promising therapeutic strategy. This review discusses the current understanding of sphingolipids' involvement in inflammation and cardiovascular diseases, the existing therapeutic approaches and gaps in therapy, and explores the potential of sphingolipids-based drugs as a future avenue for CVD treatment.
Collapse
Affiliation(s)
- Sapir Ya'ar Bar
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Noam Pintel
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Hesen Abd Alghne
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| | - Hamdan Khattib
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Department of Gastroenterology and Hepatology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Dorit Avni
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| |
Collapse
|
17
|
Gonzalez-Ramiro H, Gil MA, Cuello C, Cambra JM, Gonzalez-Plaza A, Vazquez JM, Vazquez JL, Rodriguez-Martinez H, Lucas-Sanchez A, Parrilla I, Martinez CA, Martinez EA. The Use of a Brief Synchronization Treatment after Weaning, Combined with Superovulation, Has Moderate Effects on the Gene Expression of Surviving Pig Blastocysts. Animals (Basel) 2023; 13:ani13091568. [PMID: 37174605 PMCID: PMC10177444 DOI: 10.3390/ani13091568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
The combination of estrus synchronization and superovulation (SS) treatments causes alterations in ovarian and endometrial gene expression patterns, resulting in abnormal follicle and oocyte growth, fertilization, and embryo development. However, the impact of combined SS treatments on the transcriptome of the surviving embryos remains unidentified. In this study, we examined gene expression changes in day 6 blastocysts that survived a brief regimen of synchronization treatment combined with superovulation. The sows were included in one of three groups: SS7 group (n = 6), sows were administered Altrenogest (ALT) 7 days from the day of weaning and superovulated with eCG 24 h after the end of ALT treatment and hCG at the onset of estrus; SO group (n = 6), ALT nontreated sows were superovulated with eCG 24 h postweaning and hCG at the onset of estrus; control group (n = 6), weaned sows displaying natural estrus. Six days after insemination, the sows underwent a surgical intervention for embryo collection. Transcriptome analysis was performed on blastocyst-stage embryos with good morphology. Differentially expressed genes (DEGs) between groups were detected using one-way ANOVA with an un-adjusted p-value < 0.05 and a fold change </> 1.5. The effect of SO treatment on the number of altered pathways and DEGs within each pathway was minimal. Only four pathways were disrupted comprising only a total of four altered transcripts, which were not related to reproductive functions or embryonic development. On the other hand, the surviving blastocysts subjected to SS7 treatments exhibited moderate gene expression changes in terms of DEGs and fold changes, with seven pathways disrupted containing a total of 10 transcripts affected. In this case, the up-regulation of certain pathways, such as the metabolic pathway, with two up-regulated genes associated with reproductive functions, namely RDH10 and SPTLC2, may suggest suboptimal embryo quality, while the down-regulation of others, such as the glutathione metabolism pathway, with down-regulated genes related to cellular detoxification of reactive oxygen species, namely GSTK1 and GSTO1, could depress the embryos' response to oxidative stress, thereby impairing subsequent embryo development. The gene expression changes observed in the present study in SS7 embryos, along with previous reports indicating SS7 can negatively affect fertilization, embryo production, and reproductive tract gene expression, make its use in embryo transfer programs unrecommendable.
Collapse
Affiliation(s)
- Henar Gonzalez-Ramiro
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
- Department of Research and Development, Grupo Agropor I+D+I, AIE, 30565 Murcia, Spain
| | - Maria A Gil
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Cristina Cuello
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Josep M Cambra
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Alejandro Gonzalez-Plaza
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Juan M Vazquez
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Jose L Vazquez
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Heriberto Rodriguez-Martinez
- Department of Biomedical & Clinical Sciences (BKV), BKH/Obstetrics & Gynecology, Faculty of Medicine and Health Sciences, Linköping University, 58185 Linköping, Sweden
| | | | - Inmaculada Parrilla
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| | - Cristina A Martinez
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA-CSIC), 28040 Madrid, Spain
| | - Emilio A Martinez
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research, University of Murcia, 30100 Murcia, Spain
- Institute for Biomedical Research of Murcia (IMIB-Pascual Parrilla), Campus de Ciencias de la Salud, 30120 Murcia, Spain
| |
Collapse
|
18
|
Njeim R, Alkhansa S, Fornoni A. Unraveling the Crosstalk between Lipids and NADPH Oxidases in Diabetic Kidney Disease. Pharmaceutics 2023; 15:pharmaceutics15051360. [PMID: 37242602 DOI: 10.3390/pharmaceutics15051360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Diabetic kidney disease (DKD) is a serious complication of diabetes mellitus and a leading cause of end-stage renal disease. Abnormal lipid metabolism and intrarenal accumulation of lipids have been shown to be strongly correlated with the development and progression of diabetic kidney disease (DKD). Cholesterol, phospholipids, triglycerides, fatty acids, and sphingolipids are among the lipids that are altered in DKD, and their renal accumulation has been linked to the pathogenesis of the disease. In addition, NADPH oxidase-induced production of reactive oxygen species (ROS) plays a critical role in the development of DKD. Several types of lipids have been found to be tightly linked to NADPH oxidase-induced ROS production. This review aims to explore the interplay between lipids and NADPH oxidases in order to provide new insights into the pathogenesis of DKD and identify more effective targeted therapies for the disease.
Collapse
Affiliation(s)
- Rachel Njeim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sahar Alkhansa
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
- AUB Diabetes, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
19
|
Burgio AL, Shrader CW, Kharel Y, Huang T, Salamoun JM, Lynch KR, Santos WL. 2-Aminobenzoxazole Derivatives as Potent Inhibitors of the Sphingosine-1-Phosphate Transporter Spinster Homolog 2 (Spns2). J Med Chem 2023; 66:5873-5891. [PMID: 37010497 PMCID: PMC10167756 DOI: 10.1021/acs.jmedchem.3c00149] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
The S1P1 receptor is the target of four marketed drugs for the treatment of multiple sclerosis and ulcerative colitis. Targeting an S1P exporter, specifically Spns2, that is "upstream" of S1P receptor engagement is an alternate strategy that might recapitulate the efficacy of S1P receptor modulators without cardiac toxicity. We recently reported the first Spns2 inhibitor SLF1081851 (16d) that has modest potency with in vivo activity. To develop more potent compounds, we initiated a structure-activity relationship study that identified 2-aminobenzoxazole as a viable scaffold. Our studies revealed SLB1122168 (33p), which is a potent inhibitor (IC50 = 94 ± 6 nM) of Spns2-mediated S1P release. Administration of 33p to mice and rats resulted in a dose-dependent decrease in circulating lymphocytes, a pharmacodynamic indication of Spns2 inhibition. 33p provides a valuable tool compound to explore both the therapeutic potential of targeting Spns2 and the physiologic consequences of selective S1P export inhibition.
Collapse
Affiliation(s)
- Ariel L. Burgio
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061
| | - Christopher W. Shrader
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Tao Huang
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Joseph M. Salamoun
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061
| | - Kevin R. Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908
| | - Webster L. Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, VA 24061
| |
Collapse
|
20
|
Zhao Y, Zhang Y, Li J, Zhang N, Jin Q, Qi Y, Song P. Pathogenic sphingosine 1-phosphate pathway in psoriasis: a critical review of its pathogenic significance and potential as a therapeutic target. Lipids Health Dis 2023; 22:52. [PMID: 37072847 PMCID: PMC10111724 DOI: 10.1186/s12944-023-01813-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/04/2023] [Indexed: 04/20/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a sphingolipid mediator that exerts a variety of biological functions, including immune, cardiovascular, and neurological regulation as well as tumor promotion, through high-affinity G protein-coupled receptors (S1P1-5). It has been reported that circulating S1P levels remain higher in patients with psoriasis than in healthy individuals and that circulating S1P levels do not decrease after anti-TNF-α treatment in those patients. The S1P-S1PR signaling system plays an important role in inhibiting keratinocyte proliferation, regulating lymphocyte migration, and promoting angiogenesis, thus contributing to the regulation of psoriasis pathogenesis. Here, we review the mechanisms by which S1P-S1PR signaling affects the development of psoriasis and the available clinical/preclinical evidence for targeting S1P-S1PR in psoriasis. S1P-S1PR signaling mechanisms may partially explain the link between psoriasis and its comorbidities. Although the detailed mechanisms remain to be elucidated, S1P may be a new target for future psoriasis remission.
Collapse
Affiliation(s)
- Yuechun Zhao
- Guang'anmen Hospital China Academy of Chinese Medical Sciences, Beijing, China
- Beijing University of Traditional Chinese Medicine, Chaoyang, China
| | - Yuheng Zhang
- Beijing University of Traditional Chinese Medicine, Chaoyang, China
| | - Jiaqi Li
- Guang'anmen Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Ningxin Zhang
- Beijing University of Traditional Chinese Medicine, Chaoyang, China
| | - Qiubai Jin
- Guang'anmen Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuxia Qi
- Beijing University of Traditional Chinese Medicine, Chaoyang, China
| | - Ping Song
- Guang'anmen Hospital China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
21
|
Menta V, Agarwal S, Das US, Moksha L, Srividya G, Anandan AM, Srinivasan B, Iyer G, Velpandian T, Angayarkanni N. Ocular surface sphingolipids associate with the refractory nature of vernal keratoconjunctivitis: newer insights in VKC pathogenesis. Br J Ophthalmol 2023; 107:461-469. [PMID: 34670752 DOI: 10.1136/bjophthalmol-2021-319324] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 10/05/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND The etiopathogenesis of vernal keratoconjunctivitis (VKC) is incompletely understood. Bioactive lipids play a key role in allergic disorders. This study focused on the sphingolipid metabolism on the ocular surface of VKC and to explore if it has a contributory role in the refractoriness of the disease. METHODS Active VKC cases, (n=87) (classified as mild/moderate and severe/very severe based on the disease symptoms) and age-matched healthy controls (n=60) were recruited as part of a 2-year prospective study at a tertiary eye care centre in South India. Conjunctival imprint cytology was used to assess gene expression of enzymes of sphingolipids metabolism. Sphingolipids were estimated in the tears by LC-MS/MS analysis. In vitro study was done to assess IgE-induced alterations in sphingosine-1-phosphate (S1P) receptor expression and histone modification in cultured mast cells. RESULTS Significantly altered gene expression of the sphingolipids enzymes and S1P receptor (SIP3R) were observed in conjunctival imprint cells of VKC cases. Pooled tears analysis revealed significantly lowered levels of S1P(d17:0), S1P(d20:1) (p<0.001) and S1P(d17:1) (p<0.01) specifically in severe/very severe VKC compared with both mild/moderate VKC and control. Cer(d18:/17:0) (p<0.001), ceramide-1-phosphate (C1P)(d18:1/8:0) (p<0.01) and C1P(d18:1/2.0 (p<0.05) were lowered in severe/very severe VKC compared with mild/moderate VKC. Cultured mast cells treated with IgE revealed significantly increased gene expression of S1P1 and 3 receptors and the protein expression of histone deacetylases (1, 6). CONCLUSION Altered sphingolipid metabolism in the ocular surface results in low tear ceramide and sphingosine levels in severe/very severe VKC compared with the mild/moderate cases. The novel finding opens up fresh targets for intervention in these refractory cases.
Collapse
Affiliation(s)
- Vignesh Menta
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, Vision Research Foundation, Chennai, Tamil Nadu, India
| | - Shweta Agarwal
- C.J. Shah Cornea Services, Dr G Sitalaksmi Memorial Clinic for Ocular Surface Disorders, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Ujjalkumar Subhash Das
- Ocular Pharmacology and Pharmacy Division, All India Institute of Medical Sciences, New Delhi, India
| | - Laxmi Moksha
- Ocular Pharmacology and Pharmacy Division, All India Institute of Medical Sciences, New Delhi, India
| | - Gurumurthy Srividya
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, Vision Research Foundation, Chennai, Tamil Nadu, India
| | - Amrutha M Anandan
- C.J. Shah Cornea Services, Dr G Sitalaksmi Memorial Clinic for Ocular Surface Disorders, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Bhaskar Srinivasan
- C.J. Shah Cornea Services, Dr G Sitalaksmi Memorial Clinic for Ocular Surface Disorders, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Geetha Iyer
- C.J. Shah Cornea Services, Dr G Sitalaksmi Memorial Clinic for Ocular Surface Disorders, Medical Research Foundation, Sankara Nethralaya, Chennai, Tamil Nadu, India
| | - Thirumurthy Velpandian
- Ocular Pharmacology and Pharmacy Division, All India Institute of Medical Sciences, New Delhi, India
| | - Narayanasamy Angayarkanni
- R.S. Mehta Jain Department of Biochemistry and Cell Biology, Vision Research Foundation, Chennai, Tamil Nadu, India
| |
Collapse
|
22
|
Cunha LC, Barreto LP, Valadares VS, Oliveira CFB, Vuitika L, Vilela MP, Cino EA, Silva AHDM, Nagem RAP, Chávez-Olórtegui C, Dias-Lopes C, Molina F, Felicori L. The C-terminal mutation beyond the catalytic site of brown spider phospholipase D significantly impacts its biological activities. Biochimie 2023; 211:122-130. [PMID: 36963559 DOI: 10.1016/j.biochi.2023.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/16/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023]
Abstract
Loxosceles spider envenomation results in dermonecrosis, principally due to phospholipases D (PLDs) present in the venom. These enzymes have a strongly conserved sequence, 273ATXXDNPW280, in the C-terminal region (SMD-tail) that make contact with β-sheets of the TIM barrel, in which the amino acids Asp277 and Trp280 establish the energetically strongest contacts. The SMD-tail is conserved in PLDs from different species but absent in the non-toxic PLD ancestral glycerophosphodiester phosphodiesterases (GDPDs). This work aims to understand the role of the C-terminal region in the structural stability and/or function of phospholipases D. Through site-directed mutagenesis of the rLiD1 protein (recombinant Loxosceles intermedia dermonecrotic protein 1), we produced two mutants: rLiD1D277A and rLiD1W280A (both with sphingomyelinase activity), in which Asp277 and Trp280 were replaced by alanine. rLiD1D277A showed similar sphingomyelinase activity but at least 2 times more dermonecrotic activity than rLiD1 (wild-type protein). Conversely, while the rLiD1W280A displayed a slight increase in sphingomyelinase activity, its biological activity was similar or lower compared to rLiD1, potentially due to its decreased thermostability and formation of amyloid aggregates. In conclusion, these new findings provide evidence that SMD-tail mutants impact the structure and function of these proteins and point out that residues outside the active site can even increase the function of these enzymes.
Collapse
Affiliation(s)
- Laís Cardoso Cunha
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas - ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Passos Barreto
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas - ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Veronica Silva Valadares
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas - ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila Franco Batista Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas - ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Larissa Vuitika
- Department of Immunology, Institute of Biomedical Sciences IV, University of São Paulo (ICB-IV/USP), São Paulo, Brazil
| | - Maura Páscoa Vilela
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas - ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Elio A Cino
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas - ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Ronaldo A P Nagem
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas - ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Carlos Chávez-Olórtegui
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas - ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila Dias-Lopes
- Colégio Técnico, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Franck Molina
- CNRS, SYS2DIAG-ALCEDIAG, Cap Delta, 1682 Rue de La Valsière, 34184, Montpellier, France
| | - Liza Felicori
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas - ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
23
|
Ge J, Koutarapu S, Jha D, Dulewicz M, Zetterberg H, Blennow K, Hanrieder J. Tetramodal Chemical Imaging Delineates the Lipid-Amyloid Peptide Interplay at Single Plaques in Transgenic Alzheimer's Disease Models. Anal Chem 2023; 95:4692-4702. [PMID: 36856542 PMCID: PMC10018455 DOI: 10.1021/acs.analchem.2c05302] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/16/2023] [Indexed: 03/02/2023]
Abstract
Beta-amyloid (Aβ) plaque pathology is one of the most prominent histopathological feature of Alzheimer's disease (AD). The exact pathogenic mechanisms linking Aβ to AD pathogenesis remain however not fully understood. Recent advances in amyloid-targeting pharmacotherapies highlight the critical relevance of Aβ aggregation for understanding the molecular basis of AD pathogenesis. We developed a novel, integrated, tetramodal chemical imaging paradigm for acquisition of trimodal mass spectrometry imaging (MSI) and interlaced fluorescent microscopy from a single tissue section. We used this approach to comprehensively investigate lipid-Aβ correlates at single plaques in two different mouse models of AD (tgAPPSwe and tgAPPArcSwe) with varying degrees of intrinsic properties affecting amyloid aggregation. Integration of the multimodal imaging data and multivariate data analysis identified characteristic patterns of plaque-associated lipid- and peptide localizations across both mouse models. Correlative fluorescence microscopy using structure-sensitive amyloid probes identified intra-plaque structure-specific lipid- and Aβ patterns, including Aβ 1-40 and Aβ 1-42 along with gangliosides (GM), phosphoinositols (PI), conjugated ceramides (CerP and PE-Cer), and lysophospholipids (LPC, LPA, and LPI). Single plaque correlation analysis across all modalities further revealed how these distinct lipid species were associated with Aβ peptide deposition across plaque heterogeneity, indicating different roles for those lipids in plaque growth and amyloid fibrillation, respectively. Here, conjugated ceramide species correlated with Aβ core formation indicating their involvement in initial plaque seeding or amyloid maturation. In contrast, LPI and PI were solely correlated with general plaque growth. In addition, GM1 and LPC correlated with continuous Aβ deposition and maturation. The results highlight the potential of this comprehensive multimodal imaging approach and implement distinct lipids in amyloidogenic proteinopathy.
Collapse
Affiliation(s)
- Junyue Ge
- Department
of Psychiatry and Neurochemistry, Sahlgrenska
Academy at the University of Gothenburg, Mölndal Hospital, House V3, SE-431 80 Mölndal, Sweden
| | - Srinivas Koutarapu
- Department
of Psychiatry and Neurochemistry, Sahlgrenska
Academy at the University of Gothenburg, Mölndal Hospital, House V3, SE-431 80 Mölndal, Sweden
| | - Durga Jha
- Department
of Psychiatry and Neurochemistry, Sahlgrenska
Academy at the University of Gothenburg, Mölndal Hospital, House V3, SE-431 80 Mölndal, Sweden
| | - Maciej Dulewicz
- Department
of Psychiatry and Neurochemistry, Sahlgrenska
Academy at the University of Gothenburg, Mölndal Hospital, House V3, SE-431 80 Mölndal, Sweden
| | - Henrik Zetterberg
- Department
of Psychiatry and Neurochemistry, Sahlgrenska
Academy at the University of Gothenburg, Mölndal Hospital, House V3, SE-431 80 Mölndal, Sweden
- Clinical
Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal
Hospital, House V3, SE-431 80 Mölndal, Sweden
- Department
of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
- UK
Dementia Research Institute at University College London, Queen Square, London WC1N 3BG, United Kingdom
- Hong
Kong Center for Neurodegenerative Diseases, Hong Kong 1512-1518, China
- Wisconsin
Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University
of Wisconsin-Madison, Madison, Wisconsin 53726, United States
| | - Kaj Blennow
- Department
of Psychiatry and Neurochemistry, Sahlgrenska
Academy at the University of Gothenburg, Mölndal Hospital, House V3, SE-431 80 Mölndal, Sweden
- Clinical
Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal
Hospital, House V3, SE-431 80 Mölndal, Sweden
| | - Jörg Hanrieder
- Department
of Psychiatry and Neurochemistry, Sahlgrenska
Academy at the University of Gothenburg, Mölndal Hospital, House V3, SE-431 80 Mölndal, Sweden
- Clinical
Neurochemistry Laboratory, Sahlgrenska University
Hospital, Mölndal
Hospital, House V3, SE-431 80 Mölndal, Sweden
- Department
of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London WC1N 3BG, United Kingdom
| |
Collapse
|
24
|
Yamazaki A, Kawashima A, Honda T, Kohama T, Murakami C, Sakane F, Murayama T, Nakamura H. Identification and characterization of diacylglycerol kinase ζ as a novel enzyme producing ceramide-1-phosphate. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159307. [PMID: 36906254 DOI: 10.1016/j.bbalip.2023.159307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/16/2023] [Accepted: 03/01/2023] [Indexed: 03/13/2023]
Abstract
Ceramide-1-phosphate (C1P) is a sphingolipid formed by the phosphorylation of ceramide; it regulates various physiological functions, including cell survival, proliferation, and inflammatory responses. In mammals, ceramide kinase (CerK) is the only C1P-producing enzyme currently known. However, it has been suggested that C1P is also produced by a CerK-independent pathway, although the identity of this CerK-independent C1P was unknown. Here, we identified human diacylglycerol kinase (DGK) ζ as a novel C1P-producing enzyme and demonstrated that DGKζ catalyzes the phosphorylation of ceramide to produce C1P. Analysis using fluorescently labeled ceramide (NBD-ceramide) demonstrated that only DGKζ among ten kinds of DGK isoforms increased C1P production by transient overexpression of the DGK isoforms. Furthermore, an enzyme activity assay using purified DGKζ revealed that DGKζ could directly phosphorylate ceramide to produce C1P. Furthermore, genetic deletion of DGKζ decreased the formation of NBD-C1P and the levels of endogenous C18:1/24:1- and C18:1/26:0-C1P. Interestingly, the levels of endogenous C18:1/26:0-C1P were not decreased by the knockout of CerK in the cells. These results suggest that DGKζ is also involved in the formation of C1P under physiological conditions.
Collapse
Affiliation(s)
- Ayako Yamazaki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Ayane Kawashima
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Takuya Honda
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Takafumi Kohama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Chiaki Murakami
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan; Institute for Advanced Academic Research, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Toshihiko Murayama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Hiroyuki Nakamura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan.
| |
Collapse
|
25
|
Oizumi H, Sugimura Y, Totsune T, Kawasaki I, Ohshiro S, Baba T, Kimpara T, Sakuma H, Hasegawa T, Kawahata I, Fukunaga K, Takeda A. Plasma sphingolipid abnormalities in neurodegenerative diseases. PLoS One 2022; 17:e0279315. [PMID: 36525454 PMCID: PMC9757566 DOI: 10.1371/journal.pone.0279315] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In recent years, there has been increasing evidence that several lipid metabolism abnormalities play an important role in the pathogenesis of neurodegenerative diseases. However, it is still unclear which lipid metabolism abnormalities play the most important role in neurodegenerative diseases. Plasma lipid metabolomics (lipidomics) has been shown to be an unbiased method that can be used to explore lipid metabolism abnormalities in neurodegenerative diseases. Plasma lipidomics in neurodegenerative diseases has been performed only in idiopathic Parkinson's disease (IPD) and Alzheimer's disease (AD), and comprehensive studies are needed to clarify the pathogenesis. METHODS In this study, we investigated plasma lipids using lipidomics in individuals with neurodegenerative diseases and healthy controls (CNs). Plasma lipidomics was evaluated by liquid chromatography-tandem mass spectrometry (LC-MS/MS) in those with IPD, dementia with Lewy bodies (DLB), multiple system atrophy (MSA), AD, and progressive supranuclear palsy (PSP) and CNs. RESULTS The results showed that (1) plasma sphingosine-1-phosphate (S1P) was significantly lower in all neurodegenerative disease groups (IPD, DLB, MSA, AD, and PSP) than in the CN group. (2) Plasma monohexylceramide (MonCer) and lactosylceramide (LacCer) were significantly higher in all neurodegenerative disease groups (IPD, DLB, MSA, AD, and PSP) than in the CN group. (3) Plasma MonCer levels were significantly positively correlated with plasma LacCer levels in all enrolled groups. CONCLUSION S1P, Glucosylceramide (GlcCer), the main component of MonCer, and LacCer are sphingolipids that are biosynthesized from ceramide. Recent studies have suggested that elevated GlcCer and decreased S1P levels in neurons are related to neuronal cell death and that elevated LacCer levels induce neurodegeneration by neuroinflammation. In the present study, we found decreased plasma S1P levels and elevated plasma MonCer and LacCer levels in those with neurodegenerative diseases, which is a new finding indicating the importance of abnormal sphingolipid metabolism in neurodegeneration.
Collapse
Affiliation(s)
- Hideki Oizumi
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Yoko Sugimura
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Tomoko Totsune
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Iori Kawasaki
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Saki Ohshiro
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Toru Baba
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Teiko Kimpara
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Hiroaki Sakuma
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Takafumi Hasegawa
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ichiro Kawahata
- Department of Pharmacology, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Atsushi Takeda
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
- Department of Cognitive and Motor Aging, Tohoku University Graduate School of Medicine, Sendai, Japan
- * E-mail:
| |
Collapse
|
26
|
Rybova J, Kuchar L, Sikora J, McKillop WM, Medin JA. Skin inflammation and impaired adipogenesis in a mouse model of acid ceramidase deficiency. J Inherit Metab Dis 2022; 45:1175-1190. [PMID: 36083604 PMCID: PMC9826362 DOI: 10.1002/jimd.12552] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 08/29/2022] [Accepted: 09/06/2022] [Indexed: 01/11/2023]
Abstract
Acid ceramidase catalyzes the degradation of ceramide into sphingosine and a free fatty acid. Acid ceramidase deficiency results in lipid accumulation in many tissues and leads to the development of Farber disease (FD). Typical manifestations of classical FD include formation of subcutaneous nodules and joint contractures as well as the development of a hoarse voice. Healthy skin depends on a unique lipid profile to form a barrier that confers protection from pathogens, prevents excessive water loss, and mediates cell-cell communication. Ceramides comprise ~50% of total epidermis lipids and regulate cutaneous homeostasis and inflammation. Abnormal skin development including visual skin lesions has been reported in FD patients, but a detailed study of FD skin has not been performed. We conducted a pathophysiological study of the skin in our mouse model of FD. We observed altered lipid composition in FD skin dominated by accumulation of all studied ceramide species and buildup of abnormal storage structures affecting mainly the dermis. A deficiency of acid ceramidase activity also led to the activation of inflammatory IL-6/JAK/signal transducer and activator of transcription 3 and noncanonical NF-κB signaling pathways. Last, we report reduced proliferation of FD mouse fibroblasts and adipose-derived stem/stromal cells (ASC) along with impaired differentiation of ASCs into mature adipocytes.
Collapse
Affiliation(s)
- Jitka Rybova
- Departments of Pediatrics and BiochemistryMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Ladislav Kuchar
- Rare Diseases Research Unit, Department of Pediatrics and Inherited Metabolic DisordersCharles University, 1st Faculty of Medicine and General University HospitalPragueCzech Republic
| | - Jakub Sikora
- Rare Diseases Research Unit, Department of Pediatrics and Inherited Metabolic DisordersCharles University, 1st Faculty of Medicine and General University HospitalPragueCzech Republic
- Institute of PathologyCharles University, 1st Faculty of Medicine and General University HospitalPragueCzech Republic
| | - William M. McKillop
- Departments of Pediatrics and BiochemistryMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Jeffrey A. Medin
- Departments of Pediatrics and BiochemistryMedical College of WisconsinMilwaukeeWisconsinUSA
- Department of BiochemistryMedical College of WisconsinMilwaukeeWisconsinUSA
| |
Collapse
|
27
|
Abdel Baky NA, Al-Najjar AH, Elariny HA, Sallam AS, Mohammed AA. Pramipexole and Lactoferrin ameliorate Cyclophosphamide-Induced haemorrhagic cystitis via targeting Sphk1/S1P/MAPK, TLR-4/NF-κB, and NLRP3/caspase-1/IL-1β signalling pathways and modulating the Nrf2/HO-1 pathway. Int Immunopharmacol 2022; 112:109282. [DOI: 10.1016/j.intimp.2022.109282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/30/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022]
|
28
|
Abeyrathne EDNS, Nam KC, Huang X, Ahn DU. Egg yolk lipids: separation, characterization, and utilization. Food Sci Biotechnol 2022; 31:1243-1256. [PMID: 35992319 PMCID: PMC9385935 DOI: 10.1007/s10068-022-01138-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/22/2022] [Accepted: 07/12/2022] [Indexed: 11/04/2022] Open
Abstract
Egg yolk contains very high levels of lipids, which comprise 33% of whole egg yolk. Although triglyceride is the main lipid, egg yolk is the richest source of phospholipids and cholesterol in nature. The egg yolk phospholipids have a unique composition with high levels of phosphatidylcholine followed by phosphatidylethanolamine, sphingomyelin, plasmalogen, and phosphatidylinositol. All the egg yolk lipids are embedded inside the HDL and LDL micelles or granular particles. Egg yolk lipids can be easily extracted using solvents or supercritical extraction methods but their commercial applications of egg yolk lipids are limited. Egg yolk lipids have excellent potential as a food ingredient or cosmeceutical, pharmaceutical, and nutraceutical agents because they have excellent functional and biological characteristics. This review summarizes the current knowledge on egg yolk lipids' extraction methods and functions and discusses their current and future use, which will be important to increase the use and value of the egg.
Collapse
Affiliation(s)
- Edirisingha Dewage Nalaka Sandun Abeyrathne
- Department of Animal Science, Uva Wellassa University, Badulla, 90000 Sri Lanka
- Department of Animal Science & Technology, Suncheon National University, Suncheon, 57922 Korea
| | - Ki-Chang Nam
- Department of Animal Science & Technology, Suncheon National University, Suncheon, 57922 Korea
| | - Xi Huang
- Key Laboratory of Environment Correlative Dietology, Ministry of Education, National Research and Development Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei People’s Republic of China
| | - Dong Uk Ahn
- Department of Animal Science, Iowa State University, Ames, IA 50011 USA
| |
Collapse
|
29
|
Takagi Y, Nishikado S, Omi J, Aoki J. The Many Roles of Lysophospholipid Mediators and Japanese Contributions to This Field. Biol Pharm Bull 2022; 45:1008-1021. [DOI: 10.1248/bpb.b22-00304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yugo Takagi
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Shun Nishikado
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Jumpei Omi
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo
| |
Collapse
|
30
|
Changes in the lipidome of water buffalo milk during intramammary infection by non-aureus Staphylococci. Sci Rep 2022; 12:9665. [PMID: 35690599 PMCID: PMC9188581 DOI: 10.1038/s41598-022-13400-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
This study aimed to determine the lipidome of water buffalo milk with intramammary infection (IMI) by non-aureus staphylococci (NAS), also defined as coagulase-negative staphylococci, using an untargeted lipidomic approach. Non-aureus Staphylococci are the most frequently isolated pathogens from dairy water buffalo milk during mastitis. A total of 17 milk samples from quarters affected by NAS-IMI were collected, and the lipidome was determined by liquid chromatography-quadrupole time-of-flight mass spectrometry. The results were compared with the lipidome determined on samples collected from 16 healthy quarters. The study identified 1934 different lipids, which were classified into 15 classes. The abundance of 72 lipids changed in NAS-IMI milk compared to healthy quarters. Significant changes occurred primarily in the class of free fatty acids. The results of this study provided first-time insight into the lipidome of dairy water buffalo milk. Moreover, the present findings provide evidence that NAS-IMI induces changes in water buffalo milk's lipidome.
Collapse
|
31
|
Sphingosine 1-phosphate modulation and immune cell trafficking in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2022; 19:351-366. [PMID: 35165437 DOI: 10.1038/s41575-021-00574-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2021] [Indexed: 12/12/2022]
Abstract
Immune cell trafficking is a critical element of the intestinal immune response, both in homeostasis and in pathological conditions associated with inflammatory bowel disease (IBD). This process involves adhesion molecules, chemoattractants and receptors expressed on immune cell surfaces, blood vessels and stromal intestinal tissue as well as signalling pathways, including those modulated by sphingosine 1-phosphate (S1P). The complex biological processes of leukocyte recruitment, activation, adhesion and migration have been targeted by various monoclonal antibodies (vedolizumab, etrolizumab, ontamalimab). Promising preclinical and clinical data with several oral S1P modulators suggest that inhibition of lymphocyte egress from the lymph nodes to the bloodstream might be a safe and efficacious alternative mechanism for reducing inflammation in immune-mediated disorders, including Crohn's disease and ulcerative colitis. Although various questions remain, including the potential positioning of S1P modulators in treatment algorithms and their long-term safety, this novel class of compounds holds great promise. This Review summarizes the critical mediators and mechanisms involved in immune cell trafficking in IBD and the available evidence for efficacy, safety and pharmacokinetics of S1P receptor modulators in IBD and other immune-mediated disorders. Further, it discusses potential future approaches to incorporate S1P modulators into the treatment of IBD.
Collapse
|
32
|
Tran PM, Tang SS, Salgado-Pabón W. Staphylococcus aureus β-Toxin Exerts Anti-angiogenic Effects by Inhibiting Re-endothelialization and Neovessel Formation. Front Microbiol 2022; 13:840236. [PMID: 35185854 PMCID: PMC8851161 DOI: 10.3389/fmicb.2022.840236] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/11/2022] [Indexed: 12/25/2022] Open
Abstract
Staphylococcus aureus causes severe, life-threatening infections that often are complicated by severe local and systemic pathologies with non-healing lesions. A classic example is S. aureus infective endocarditis (IE), where the secreted hemolysin β-toxin potentiates the disease via its sphingomyelinase and biofilm ligase activities. Although these activities dysregulate human aortic endothelial cell activation, β-toxin effect on endothelial cell function in wound healing has not been addressed. With the use of the ex vivo rabbit aortic ring model, we provide evidence that β-toxin prevents branching microvessel formation, highlighting its ability to interfere with tissue re-vascularization and vascular repair. We show that β-toxin specifically targets both human aortic endothelial cell proliferation and cell migration and inhibits human umbilical vein endothelial cell rearrangement into capillary-like networks in vitro. Proteome arrays specific for angiogenesis-related molecules provided evidence that β-toxin promotes an inhibitory profile in endothelial cell monolayers, specifically targeting production of TIMP-1, TIMP-4, and IGFBP-3 to counter the effect of a pro-angiogenic environment. Dysregulation in the production of these molecules is known to result in sprouting defects (including deficient cell proliferation, migration, and survival), vessel instability and/or vascular regression. When endothelial cells are grown under re-endothelialization/wound healing conditions, β-toxin decreases the pro-angiogenic molecule MMP-8 and increases the anti-angiogenic molecule endostatin. Altogether, the data indicate that β-toxin is an anti-angiogenic virulence factor and highlight a mechanism where β-toxin exacerbates S. aureus invasive infections by interfering with tissue re-vascularization and vascular repair.
Collapse
Affiliation(s)
- Phuong M. Tran
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
- Department of Microbiology and Immunology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Sharon S. Tang
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Wilmara Salgado-Pabón
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
33
|
Revealing 2-Dimethylhydrazino-2-alkyl alkynyl sphingosine derivatives as Sphingosine Kinase 2 inhibitors: some hints on the structural basis for selective inhibition. Bioorg Chem 2022; 121:105668. [DOI: 10.1016/j.bioorg.2022.105668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 12/29/2022]
|
34
|
Yaobitong capsules reshape and rebalance the gut microbiota and metabolites of arthritic rats: An integrated study of microbiome and fecal metabolomics analysis. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1190:123096. [PMID: 34998201 DOI: 10.1016/j.jchromb.2021.123096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/19/2021] [Accepted: 12/30/2021] [Indexed: 12/13/2022]
Abstract
Yaobitong capsule (YBTC), a Chinese medicine compound preparation, has been demonstrated to affect multiple pathways associated with inflammation and exhibit potential anti-arthritis effect. In this study, an integrated omic approach based on UHPLC-Q-TOF MS and 16S rRNA sequencing analyses was proposed to reveal the anti-arthritis effect and possible mechanism of YBTC. The AIA rat model showed that YBTC significantly alleviated the typical symptoms of AIA rats such as weight, spleen index and pro-inflammatory cytokines. Fecal metabolomics results identified 41 differential metabolites, which mainly referred to tryptophan, bile acid and fatty acid metabolism. The gut microbiota played a crucially important role in anti-inflammatory immunity, 16S rRNA results indicated that YBTC changed the community structure and alleviated the microecological imbalance caused by rheumatoid arthritis (RA). Further ROC curve analysis demonstrated that it was reliable to identify RA by using 5 metabolites and 3 microorganisms (AUC > 0.83). In summary, it was the first time that the preventive effect of YBTC in RA was confirmed. The secretion of the microbiota-mediated metabolites was significantly improved by YBTC, through its callback effect on the disturbed gut microbiota. Thus, we have indicated a potential novel strategy for the prevention of RA via evaluation of intervention effects of YBTC on AIA rat model.
Collapse
|
35
|
Implication of Ceramide Kinase/C1P in Cancer Development and Progression. Cancers (Basel) 2022; 14:cancers14010227. [PMID: 35008391 PMCID: PMC8750078 DOI: 10.3390/cancers14010227] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 12/28/2022] Open
Abstract
Cancer cells rewire their metabolic programs to favor biological processes that promote cell survival, proliferation, and dissemination. Among this relevant reprogramming, sphingolipid metabolism provides metabolites that can favor or oppose these hallmarks of cancer. The sphingolipid ceramide 1-phosphate (C1P) and the enzyme responsible for its biosynthesis, ceramide kinase (CERK), are well established regulators of cell growth and survival in normal, as well as malignant cells through stress-regulated signaling pathways. This metabolite also promotes cell survival, which has been associated with the feedback regulation of other antitumoral sphingolipids or second messengers. C1P also regulates cancer cell invasion and migration of different types of cancer, including lung, breast, pancreas, prostate, or leukemia cells. More recently, CERK and C1P have been implicated in the control of inflammatory responses. The present review provides an updated view on the important role of CERK/C1P in the regulation of cancer cell growth, survival, and dissemination.
Collapse
|
36
|
Ding Y, Jiang Y, Zhu M, Zhu Q, He Y, Lu Y, Wang Y, Qi J, Feng Y, Huang R, Yin H, Li S, Sun Y. Follicular fluid lipidomic profiling reveals potential biomarkers of polycystic ovary syndrome: A pilot study. Front Endocrinol (Lausanne) 2022; 13:960274. [PMID: 36176459 PMCID: PMC9513192 DOI: 10.3389/fendo.2022.960274] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a heterogeneous endocrine disorder associated with multiple metabolic conditions including obesity, insulin resistance, and dyslipidemia. PCOS is the most common cause of anovulatory infertility; however, the molecular diversity of the ovarian follicle microenvironment is not fully understood. This study aimed to investigate the follicular fluid (FF) lipidomic profiles in different phenotypes of PCOS and to explore novel lipid biomarkers. METHODS A total of 25 women with PCOS and 12 women without PCOS who underwent in vitro fertilization and embryo transfer were recruited, and their FF samples were collected for the lipidomic study. Liquid chromatography-tandem mass spectrometry was used to compare the differential abundance of FF lipids between patients with different PCOS phenotypes and controls. Subsequently, correlations between specific lipid concentrations in FF and high-quality embryo rate (HQER) were analyzed to further evaluate the potential interferences of lipid levels with oocyte quality in PCOS. Candidate biomarkers were then compared via receiver operating characteristic (ROC) curve analysis. RESULTS In total, 19 lipids were identified in ovarian FF. Of these, the concentrations of ceramide (Cer) and free fatty acids (FFA) in FF were significantly increased, whereas those of lysophosphatidylglycerol (LPG) were reduced in women with PCOS compared to controls, especially in obese and insulin-resistant groups. In addition, six subclasses of ceramide, FFA, and LPG were correlated with oocyte quality. Twenty-three lipid subclasses were identified as potential biomarkers of PCOS, and ROC analysis indicated the prognostic value of Cer,36:1;2, FFA C14:1, and LPG,18:0 on HQER in patients with PCOS. CONCLUSIONS Our study showed the unique lipidomic profiles in FF from women with PCOS. Moreover, it provided metabolic signatures as well as candidate biomarkers that help to better understand the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Ying Ding
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yihong Jiang
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingjiang Zhu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Qinling Zhu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yaqiong He
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yao Lu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yuan Wang
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Jia Qi
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yifan Feng
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Huang
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiyong Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai, China
| | - Shengxian Li
- Department of Endocrinology and Metabolism, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Shengxian Li, ; Yun Sun,
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- *Correspondence: Shengxian Li, ; Yun Sun,
| |
Collapse
|
37
|
Wollny T, Wątek M, Wnorowska U, Piktel E, Góźdź S, Kurek K, Wolak P, Król G, Żendzian-Piotrowska M, Bucki R. Hypogelsolinemia and Decrease in Blood Plasma Sphingosine-1-Phosphate in Patients Diagnosed with Severe Acute Pancreatitis. Dig Dis Sci 2022; 67:536-545. [PMID: 33620599 PMCID: PMC8885474 DOI: 10.1007/s10620-021-06865-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 01/20/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Acute pancreatitis (AP) is a frequent hospitalization cause of patients suffering from gastrointestinal disorders. Gelsolin has an ability to bind bioactive lipids including different sphingolipids engaged in inflammatory response. Importantly, hypogelsolinemia was observed in patients with different states of acute and chronic inflammation. AIMS The aim of the present study was to assess the interplay of blood plasma gelsolin and blood plasma sphingosine-1-phosphate (S1P) concentration in patients diagnosed with acute pancreatitis. MATERIALS AND METHODS To assess the concentration of gelsolin and S1P, immunoblotting and HPLC technique were employed, respectively. Additionally, the concentrations of amylase, lipase, C-reactive protein (CRP), procalcitonin (PCT) and the number of white blood cells (WBC) and platelet (PLT) were recorded. RESULTS We found that both pGSN and S1P concentrations in the plasma of the AP patients were significantly lower (pGSN ~ 15-165 mg/L; S1P ~ 100-360 pmol/mL) when compared to the levels of pGSN and S1P in a control group (pGSN ~ 130-240 mg/L; S1P ~ 260-400 pmol/mL). Additionally, higher concentrations of CRP, WBC, amylase and lipase were associated with low level of gelsolin in the blood of AP patients. No correlations between the level of PCT and PLT with gelsolin concentration were noticed. CONCLUSION Plasma gelsolin and S1P levels decrease during severe acute pancreatitis. Simultaneous assessment of pGSN and S1P can be useful in development of more accurate diagnostic strategies for patients with severe acute pancreatitis.
Collapse
Affiliation(s)
- Tomasz Wollny
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734, Kielce, Poland
| | - Marzena Wątek
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734, Kielce, Poland
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Indiry Gandhi 14, 02-776, Warsaw, Poland
| | - Urszula Wnorowska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222, Białystok, Poland
| | - Ewelina Piktel
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222, Białystok, Poland
| | - Stanisław Góźdź
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734, Kielce, Poland
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317, Kielce, Poland
| | - Krzysztof Kurek
- Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, Skłodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Przemysław Wolak
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317, Kielce, Poland
| | - Grzegorz Król
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317, Kielce, Poland
| | - Małgorzata Żendzian-Piotrowska
- Department of Hygiene, Epidemiology and Ergonomics, Medical University of Bialystok, Mickiewicza 2C, 15-222, Bialystok, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222, Białystok, Poland.
| |
Collapse
|
38
|
Ali T, Lei X, Barbour SE, Koizumi A, Chalfant CE, Ramanadham S. Alterations in β-Cell Sphingolipid Profile Associated with ER Stress and iPLA 2β: Another Contributor to β-Cell Apoptosis in Type 1 Diabetes. Molecules 2021; 26:molecules26216361. [PMID: 34770770 PMCID: PMC8587436 DOI: 10.3390/molecules26216361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes (T1D) development, in part, is due to ER stress-induced β-cell apoptosis. Activation of the Ca2+-independent phospholipase A2 beta (iPLA2β) leads to the generation of pro-inflammatory eicosanoids, which contribute to β-cell death and T1D. ER stress induces iPLA2β-mediated generation of pro-apoptotic ceramides via neutral sphingomyelinase (NSMase). To gain a better understanding of the impact of iPLA2β on sphingolipids (SLs), we characterized their profile in β-cells undergoing ER stress. ESI/MS/MS analyses followed by ANOVA/Student’s t-test were used to assess differences in sphingolipids molecular species in Vector (V) control and iPLA2β-overexpressing (OE) INS-1 and Akita (AK, spontaneous model of ER stress) and WT-littermate (AK-WT) β-cells. As expected, iPLA2β induction was greater in the OE and AK cells in comparison with V and WT cells. We report here that ER stress led to elevations in pro-apoptotic and decreases in pro-survival sphingolipids and that the inactivation of iPLA2β restores the sphingolipid species toward those that promote cell survival. In view of our recent finding that the SL profile in macrophages—the initiators of autoimmune responses leading to T1D—is not significantly altered during T1D development, we posit that the iPLA2β-mediated shift in the β-cell sphingolipid profile is an important contributor to β-cell death associated with T1D.
Collapse
Affiliation(s)
- Tomader Ali
- Research Department, Imperial College London Diabetes Center, Abu Dhabi 51133, United Arab Emirates;
| | - Xiaoyong Lei
- Department of Cell, Developmental, and Integrative Biology and Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Suzanne E. Barbour
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Akio Koizumi
- Department of Health and Environmental Sciences, Kyoto Graduate School of Medicine, Kyoto 606-8501, Japan;
| | - Charles E. Chalfant
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL 33620, USA;
| | - Sasanka Ramanadham
- Department of Cell, Developmental, and Integrative Biology and Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Correspondence: ; Tel.: +1-205-996-5973; Fax: +1-205-996-5220
| |
Collapse
|
39
|
An JN, Kim H, Kim EN, Cho A, Cho Y, Choi YW, Kim JH, Yang SH, Choi BS, Lim CS, Kim YS, Kim KP, Lee JP. Effects of periostin deficiency on kidney aging and lipid metabolism. Aging (Albany NY) 2021; 13:22649-22665. [PMID: 34607314 PMCID: PMC8544301 DOI: 10.18632/aging.203580] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 08/31/2021] [Indexed: 11/25/2022]
Abstract
Periostin plays a crucial role in fibrosis, which is involved in kidney aging. A few studies have shown that lipid metabolism is involved in kidney aging. We investigated the role of periostin in lipid metabolism during kidney aging. Renal function, fibrosis, and inflammatory markers were studied using urine, blood, and tissue samples from wild-type (WT) C57BL/6 mice and Postn-null mice of 2 and 24 months of age. Lipids were quantitatively profiled using liquid chromatography-tandem mass spectrometry in the multiple reaction monitoring mode. Renal function was worse and tubular atrophy/interstitial fibrosis, periostin expression, and inflammatory and fibrotic markers were more severe in aged WT mice than in young WT mice. In aged Postn-null mice, these changes were mitigated. Thirty-five differentially regulated lipids were identified. Phosphatidylcholines, cholesteryl ester, cholesterol, ceramide-1-phosphate, and CCL5 expression were significantly higher in aged WT mice than in aged Postn-null mice. Particularly, linoleic acid, linolenic acid, arachidonic acid, and docosahexaenoic acid differed strongly between the two groups. Lysophosphatidylcholine acyltransferase 2, which converts lysophosphatidylcholine to phosphatidylcholine, was significantly higher in aged WT mice than in aged Postn-null mice. Periostin expression in the kidneys increased with age, and periostin ablation delayed aging. Changes in lipids and their metabolism were found in Postn-null mice. Further research on the precise mechanisms of and relationships between lipid expression and metabolism, kidney aging, and periostin expression is warranted.
Collapse
Affiliation(s)
- Jung Nam An
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, Gyeonggi-do, Korea
| | - Hyoseon Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Korea
| | - Eun Nim Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ara Cho
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Yeongeun Cho
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, Korea
| | - Young Wook Choi
- Department of Urology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Jin Hyuk Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Seung Hee Yang
- Seoul National University Kidney Research Institute, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Bum Soon Choi
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
40
|
Allaway D, Harrison M, Haydock R, Watson P. Adaptations Supporting Plasma Methionine on a Limited-Methionine, High-Cystine Diet Alter the Canine Plasma Metabolome Consistent with Interventions that Extend Life Span in Other Species. J Nutr 2021; 151:3125-3136. [PMID: 34224573 DOI: 10.1093/jn/nxab204] [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: 09/02/2020] [Revised: 09/29/2020] [Accepted: 06/03/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Using indicator amino acid oxidation methodology, the mean dietary requirement of adult dogs for methionine (Met) was estimated to be ∼66% of the current recommended allowance. Dogs fed a diet formulated to provide the estimated mean Met requirement for 32 wk maintained plasma Met, seemingly supported by betaine oxidation. OBJECTIVE To gain a better understanding of the metabolic changes that were associated with supporting plasma Met when dogs were fed a limited Met diet over 32 wk, we analyzed plasma samples taken from that study using a data-driven metabolomics approach. METHODS Labrador retrievers (20 females/13 males; mean age: 4.9 y; range: 2.0-7.9 y) were fed semi-purified, nutritionally complete diets. After 4 wk of feeding a control diet (DL-Met; 1.37 g/1000 kcal), 17 dogs remained on this diet and 16 were transitioned to a test diet formulated to the estimated mean Met requirement (0.55 g/1000 kcal), with dietary total sulfur amino acid maintained with additional l-cystine (Cys). Dogs were individually fed diets to maintain a stable body weight at an ideal body condition score for 32 wk. Plasma samples from fasted blood collected at baseline and 8 and 32 wk were analyzed using untargeted metabolic profiling. RESULTS Analysis of metabolites (n = 593) confirmed our primary findings (increased Met, betaine, and dimethylglycine). Metabolite changes consistent with repartitioning choline to support Met cycling included reduced pools of lipids derived via phosphatidylethanolamine N-methyltransferase and enhanced fatty acid oxidation. Some changes were consistent with metabolomics studies reported in other species that used interventions known to extend life span (caloric- and Met-restricted diets or feeding strategy). CONCLUSIONS Changes in the plasma metabolome were consistent with reported adaptations to support Met-dependent activities. We propose that feeding a limited-Met, high-Cys diet using the estimated mean Met requirement in adult Labrador retrievers alters regulation of the Met cycle, thereby altering metabolism, similar to interventions that extend life span.
Collapse
Affiliation(s)
- David Allaway
- WALTHAM Petcare Science Institute, Melton Mowbray, Leicestershire, United Kingdom
| | - Matthew Harrison
- WALTHAM Petcare Science Institute, Melton Mowbray, Leicestershire, United Kingdom
| | - Richard Haydock
- WALTHAM Petcare Science Institute, Melton Mowbray, Leicestershire, United Kingdom
| | - Phillip Watson
- WALTHAM Petcare Science Institute, Melton Mowbray, Leicestershire, United Kingdom
| |
Collapse
|
41
|
Xu H, Tamrat NE, Gao J, Xu J, Zhou Y, Zhang S, Chen Z, Shao Y, Ding L, Shen B, Wei Z. Combined Signature of the Urinary Microbiome and Metabolome in Patients With Interstitial Cystitis. Front Cell Infect Microbiol 2021; 11:711746. [PMID: 34527602 PMCID: PMC8436771 DOI: 10.3389/fcimb.2021.711746] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/29/2021] [Indexed: 02/02/2023] Open
Abstract
Interstitial cystitis (IC) is a clinical syndrome characterized by frequency, urgency, and bladder pain or pelvic pain; however, the underlying pathophysiological mechanisms and diagnostic markers are unknown. In this study, microbiome and metabolome analysis were used to explain the urine signatures of IC patients. Urine samples from 20 IC patients and 22 control groups were analyzed by using 16S rRNA sequence and liquid chromatography coupled with mass spectrometry. Four opportunistic pathogen genera, including Serratia, Brevibacterium, Porphyromonas, and Citrobacter, were significantly upregulated in IC group. The altered metabolite signatures of the metabolome may be related to sphingosine metabolism, amino acid metabolism, and fatty acid biosynthesis. Meanwhile, the associations were observed between different metabolites and microbiomes of IC. The present study suggests that the combined signatures of IC in urine microbiome and metabolome may become its prospective diagnostic markers.
Collapse
Affiliation(s)
- Hewei Xu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Nebiyu Elias Tamrat
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Jie Gao
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Jie Xu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Yiduo Zhou
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Sicong Zhang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Zhengsen Chen
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Yunpeng Shao
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Liucheng Ding
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Baixin Shen
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Zhongqing Wei
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Zhang YM. Orosomucoid-like protein 3, rhinovirus and asthma. World J Crit Care Med 2021; 10:170-182. [PMID: 34616654 PMCID: PMC8462028 DOI: 10.5492/wjccm.v10.i5.170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/16/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
The genetic variants of orosomucoid-like protein 3 (ORMDL3) gene are associated with highly significant increases in the number of human rhinovirus (HRV)-induced wheezing episodes in children. Recent investigations have been focused on the mechanisms of ORMDL3 in rhinovirus infection for asthma and asthma exacerbations. ORMDL3 not only regulates major human rhinovirus receptor intercellular adhesion molecule 1 expression, but also plays pivotal roles in viral infection through metabolisms of ceramide and sphingosine-1-phosphate, endoplasmic reticulum (ER) stress, ER-Golgi interface and glycolysis. Research on the roles of ORMDL3 in HRV infection will lead us to identify new biomarkers and novel therapeutic targets in childhood asthma and viral induced asthma exacerbations.
Collapse
Affiliation(s)
- You-Ming Zhang
- Section of Genomic and Environmental Medicine, National Heart and Lung Institute, Molecular Genetics Group, Division of Respiratory Sciences, Imperial College London, London SW3 6LY, United Kingdom
| |
Collapse
|
43
|
Paudel YN, Angelopoulou E, Piperi C, Gnatkovsky V, Othman I, Shaikh MF. From the Molecular Mechanism to Pre-clinical Results: Anti-epileptic Effects of Fingolimod. Curr Neuropharmacol 2021; 18:1126-1137. [PMID: 32310049 PMCID: PMC7709153 DOI: 10.2174/1570159x18666200420125017] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/13/2020] [Accepted: 04/14/2020] [Indexed: 02/08/2023] Open
Abstract
Epilepsy is a devastating neurological condition characterized by long-term tendency to generate unprovoked seizures, affecting around 1-2% of the population worldwide. Epilepsy is a serious health concern which often associates with other neurobehavioral comorbidities that further worsen disease conditions. Despite tremendous research, the mainstream anti-epileptic drugs (AEDs) exert only symptomatic relief leading to 30% of untreatable patients. This reflects the complexity of the disease pathogenesis and urges the precise understanding of underlying mechanisms in order to explore novel therapeutic strategies that might alter the disease progression as well as minimize the epilepsy-associated comorbidities. Unfortunately, the development of novel AEDs might be a difficult process engaging huge funds, tremendous scientific efforts and stringent regulatory compliance with a possible chance of end-stage drug failure. Hence, an alternate strategy is drug repurposing, where anti-epileptic effects are elicited from drugs that are already used to treat non-epileptic disorders. Herein, we provide evidence of the anti-epileptic effects of Fingolimod (FTY720), a modulator of sphingosine-1-phosphate (S1P) receptor, USFDA approved already for Relapsing-Remitting Multiple Sclerosis (RRMS). Emerging experimental findings suggest that Fingolimod treatment exerts disease-modifying anti-epileptic effects based on its anti-neuroinflammatory properties, potent neuroprotection, anti-gliotic effects, myelin protection, reduction of mTOR signaling pathway and activation of microglia and astrocytes. We further discuss the underlying molecular crosstalk associated with the anti-epileptic effects of Fingolimod and provide evidence for repurposing Fingolimod to overcome the limitations of current AEDs.
Collapse
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vadym Gnatkovsky
- Unit of Epileptology and Experimental Neurophysiology, Fondazione Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
44
|
Regulation of cell growth, survival and migration by ceramide 1-phosphate - implications in lung cancer progression and inflammation. Cell Signal 2021; 83:109980. [PMID: 33727076 DOI: 10.1016/j.cellsig.2021.109980] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 01/10/2023]
Abstract
Ceramide 1-phosphate (C1P) is a bioactive sphingolipid that is implicated in the regulation of vital cellular functions and plays key roles in a number of inflammation-associated pathologies. C1P was first described as mitogenic for fibroblasts and macrophages and was later found to promote cell survival in different cell types. The mechanisms involved in the mitogenic actions of C1P include activation of MEK/ERK1-2, PI3K/Akt/mTOR, or PKC-α, whereas promotion of cell survival required a substantial reduction of ceramide levels through inhibition of serine palmitoyl transferase or sphingomyelinase activities. C1P and ceramide kinase (CerK), the enzyme responsible for its biosynthesis in mammalian cells, play key roles in tumor promotion and dissemination. CerK-derived C1P can be secreted to the extracellular milieu by different cell types and is also present in extracellular vesicles. In this context, whilst cell proliferation is regulated by intracellularly generated C1P, stimulation of cell migration/invasion requires the intervention of exogenous C1P. Regarding inflammation, C1P was first described as pro-inflammatory in a variety of cell types. However, cigarette smoke- or lipopolysaccharide-induced lung inflammation in mouse or human cells was overcome by pretreatment with natural or synthetic C1P analogs. Both acute and chronic lung inflammation, and the development of lung emphysema were substantially reduced by exogenous C1P applications, pointing to an anti-inflammatory action of C1P in the lungs. The molecular mechanisms involved in the regulation of cell growth, survival and migration with especial emphasis in the control of lung cancer biology are discussed.
Collapse
|
45
|
The Phenoxyphenol Compound diTFPP Mediates Exogenous C 2-Ceramide Metabolism, Inducing Cell Apoptosis Accompanied by ROS Formation and Autophagy in Hepatocellular Carcinoma Cells. Antioxidants (Basel) 2021; 10:antiox10030394. [PMID: 33807856 PMCID: PMC7998835 DOI: 10.3390/antiox10030394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 01/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a severe disease that accounts for 80% of liver cancers. Chemotherapy is the primary therapeutic strategy for patients who cannot be treated with surgery or who have late-stage HCC. C2-ceramide is an effective reagent that has been found to inhibit the growth of many cancer types. The metabolism of C2-ceramide plays a vital role in the regulation of cell death/cell survival. The phenoxyphenol compound 4-{2,3,5,6-tetrafluoro-4-[2,3,5,6-tetrafluoro-4-(4-hydroxyphenoxy)phenyl]phenoxy}phenol (diTFPP) was found to have a synergistic effect with C2-ceramide, resulting in considerable cell death in the HA22T HCC cell line. diTFPP/C2-ceramide cotreatment induced a two- to threefold increase in cell death compared to that with C2-ceramide alone and induced pyknosis. Annexin V/7-aminoactinomycin D (7AAD) double staining and Western blotting indicated that apoptosis was involved in diTFPP/C2-ceramide cotreatment-mediated cell death. We next analyzed transcriptome alterations in diTFPP/C2-ceramide-cotreated HA22T cells with next-generation sequencing (NGS). The data indicated that diTFPP treatment disrupted sphingolipid metabolism, inhibited cell cycle-associated gene expression, and induced autophagy and reactive oxygen species (ROS)-responsive changes in gene expression. Additionally, we assessed the activation of autophagy with acridine orange (AO) staining and observed alterations in the expression of the autophagic proteins LC3B-II and Beclin-1, which indicated autophagy activation after diTFPP/C2-ceramide cotreatment. Elevated levels of ROS were also reported in diTFPP/C2-ceramide-treated cells, and the expression of the ROS-associated proteins SOD1, SOD2, and catalase was upregulated after diTFPP/C2-ceramide treatment. This study revealed the potential regulatory mechanism of the novel compound diTFPP in sphingolipid metabolism by showing that it disrupts ceramide metabolism and apoptotic sphingolipid accumulation.
Collapse
|
46
|
Lu JW, Tseng YS, Lo YS, Lin YM, Yeh CM, Lin SH. Prognostic Significance of Cytoplasmic SPNS2 Expression in Patients with Oral Squamous Cell Carcinoma. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:164. [PMID: 33673355 PMCID: PMC7917906 DOI: 10.3390/medicina57020164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/09/2021] [Accepted: 02/09/2021] [Indexed: 01/09/2023]
Abstract
Background and Objectives: Oral squamous cell carcinoma (OSCC) is a malignant disease with a particularly high incidence in Taiwan. Our objective in this study was to elucidate the involvement of sphingolipid transporter 2 (SPNS2) expression and SPNS2 protein expression in the clinicopathological indexes and the clinical outcomes of OSCC patients. Materials and Methods: Immunohistochemistry analysis was performed for SPNS2 protein expression in samples from 264 cases of OSCC. Correlations of SPNS2 expression with clinicopathological variables and patient survival were analyzed. Results: Our results revealed that the cytoplasmic protein expression of SPNS2 in OSCC tissue specimens was lower than in normal tissue specimens. Negative cytoplasmic protein expression of SPNS2 was significantly correlated with T status and stage. Kaplan-Meier survival curve analysis revealed that negative cytoplasmic SPNS2 expression was predictive of poorer overall survival of OSCC patients in stage III/IV. We also determined that low SPNS2 expression was an independent prognostic factor related to overall survival among OSCC patients in stage III/IV from univariate Cox proportional hazard models. Multivariate Cox proportional hazard models revealed that cytoplasmic SPNS2 expression, T status, lymph node metastasis, and histological grade were independent prognostic factors for survival. Conclusions: Overall, this study determined that SPNS2 protein may be a useful prognostic marker for OSCC patients and potential therapeutic target for OSCC treatment.
Collapse
Affiliation(s)
- Jeng-Wei Lu
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore;
| | - Yen-Shuo Tseng
- Department of Dermatology, Changhua Christian Hospital, Changhua 500, Taiwan;
| | - Yu-Sheng Lo
- Department of Oral Cancer Research Center, Changhua Christian Hospital, Changhua 500, Taiwan;
| | - Yueh-Min Lin
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan;
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua 500, Taiwan;
| | - Chung-Min Yeh
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua 500, Taiwan;
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
| | - Shu-Hui Lin
- Department of Surgical Pathology, Changhua Christian Hospital, Changhua 500, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, Central Taiwan University of Science and Technology, Taichung 406, Taiwan
| |
Collapse
|
47
|
Role of bioactive sphingolipids in physiology and pathology. Essays Biochem 2021; 64:579-589. [PMID: 32579188 DOI: 10.1042/ebc20190091] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/27/2022]
Abstract
Sphingolipids are a class of complex lipids containing a backbone of sphingoid bases, namely the organic aliphatic amino alcohol sphingosine (Sph), that are essential constituents of eukaryotic cells. They were first described as major components of cell membrane architecture, but it is now well established that some sphingolipids are bioactive and can regulate key biological functions. These include cell growth and survival, cell differentiation, angiogenesis, autophagy, cell migration, or organogenesis. Furthermore, some bioactive sphingolipids are implicated in pathological processes including inflammation-associated illnesses such as atherosclerosis, rheumatoid arthritis, inflammatory bowel disease (namely Crohn's disease and ulcerative colitis), type II diabetes, obesity, and cancer. A major sphingolipid metabolite is ceramide, which is the core of sphingolipid metabolism and can act as second messenger, especially when it is produced at the plasma membrane of cells. Ceramides promote cell cycle arrest and apoptosis. However, ceramide 1-phosphate (C1P), the product of ceramide kinase (CerK), and Sph 1-phosphate (S1P), which is generated by the action of Sph kinases (SphK), stimulate cell proliferation and inhibit apoptosis. Recently, C1P has been implicated in the spontaneous migration of cells from some types of cancer, and can enhance cell migration/invasion of malignant cells through interaction with a Gi protein-coupled receptor. In addition, CerK and SphK are implicated in inflammatory responses, some of which are associated with cancer progression and metastasis. Hence, targeting these sphingolipid kinases to inhibit C1P or S1P production, or blockade of their receptors might contribute to the development of novel therapeutic strategies to reduce metabolic alterations and disease.
Collapse
|
48
|
Simon MV, Basu SK, Qaladize B, Grambergs R, Rotstein NP, Mandal N. Sphingolipids as critical players in retinal physiology and pathology. J Lipid Res 2021; 62:100037. [PMID: 32948663 PMCID: PMC7933806 DOI: 10.1194/jlr.tr120000972] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/04/2020] [Indexed: 12/24/2022] Open
Abstract
Sphingolipids have emerged as bioactive lipids involved in the regulation of many physiological and pathological processes. In the retina, they have been established to participate in numerous processes, such as neuronal survival and death, proliferation and migration of neuronal and vascular cells, inflammation, and neovascularization. Dysregulation of sphingolipids is therefore crucial in the onset and progression of retinal diseases. This review examines the involvement of sphingolipids in retinal physiology and diseases. Ceramide (Cer) has emerged as a common mediator of inflammation and death of neuronal and retinal pigment epithelium cells in animal models of retinopathies such as glaucoma, age-related macular degeneration (AMD), and retinitis pigmentosa. Sphingosine-1-phosphate (S1P) has opposite roles, preventing photoreceptor and ganglion cell degeneration but also promoting inflammation, fibrosis, and neovascularization in AMD, glaucoma, and pro-fibrotic disorders. Alterations in Cer, S1P, and ceramide 1-phosphate may also contribute to uveitis. Notably, use of inhibitors that either prevent Cer increase or modulate S1P signaling, such as Myriocin, desipramine, and Fingolimod (FTY720), preserves neuronal viability and retinal function. These findings underscore the relevance of alterations in the sphingolipid metabolic network in the etiology of multiple retinopathies and highlight the potential of modulating their metabolism for the design of novel therapeutic approaches.
Collapse
Affiliation(s)
- M Victoria Simon
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina
| | - Sandip K Basu
- Departments of Ophthalmology and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Bano Qaladize
- Departments of Ophthalmology and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Richard Grambergs
- Departments of Ophthalmology and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nora P Rotstein
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina.
| | - Nawajes Mandal
- Departments of Ophthalmology and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
49
|
Paradiso E, Lazzaretti C, Sperduti S, Antoniani F, Fornari G, Brigante G, Di Rocco G, Tagliavini S, Trenti T, Morini D, Falbo AI, Villani MT, Nofer JR, Simoni M, Potì F, Casarini L. Sphingosine-1 phosphate induces cAMP/PKA-independent phosphorylation of the cAMP response element-binding protein (CREB) in granulosa cells. Mol Cell Endocrinol 2021; 520:111082. [PMID: 33189864 DOI: 10.1016/j.mce.2020.111082] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/20/2020] [Accepted: 11/09/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS Sphingosine-1 phosphate (S1P) is a lysosphingolipid present in the ovarian follicular fluid. The role of the lysosphingolipid in gonads of the female is widely unclear. At nanomolar concentrations, S1P binds and activates five specific G protein-coupled receptors (GPCRs), known as S1P1-5, modulating different signaling pathways. S1P1 and S1P3 are highly expressed in human primary granulosa lutein cells (hGLC), as well as in the immortalized human primary granulosa cell line hGL5. In this study, we evaluated the signaling cascade activated by S1P and its synthetic analogues in hGLC and hGL5 cells, exploring the biological relevance of S1PR-stimulation in this context. METHODS AND RESULTS hGLC and hGL5 cells were treated with a fixed dose (0.1 μM) of S1P, or by S1P1- and S1P3-specific agonists SEW2871 and CYM5541. In granulosa cells, S1P and, at a lesser extent, SEW2871 and CYM5541, potently induced CREB phosphorylation. No cAMP production was detected and pCREB activation occurred even in the presence of the PKA inhibitor H-89. Moreover, S1P-dependent CREB phosphorylation was dampened by the mitogen-activate protein kinase (MEK) inhibitor U0126 and by the L-type Ca2+ channel blocker verapamil. The complete inhibition of CREB phosphorylation occurred by blocking either S1P2 or S1P3 with the specific receptor antagonists JTE-013 and TY52156, or under PLC/PI3K depletion. S1P-dependent CREB phosphorylation induced FOXO1 and the EGF-like epiregulin-encoding gene (EREG), confirming the exclusive role of gonadotropins and interleukins in this process, but did not affect steroidogenesis. However, S1P or agonists did not modulate granulosa cell viability and proliferation in our conditions. CONCLUSIONS This study demonstrates for the first time that S1P may induce a cAMP-independent activation of pCREB in granulosa cells, although this is not sufficient to induce intracellular steroidogenic signals and progesterone synthesis. S1P-induced FOXO1 and EREG gene expression suggests that the activation of S1P-S1PR axis may cooperate with gonadotropins in modulating follicle development.
Collapse
Affiliation(s)
- Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - Clara Lazzaretti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - Samantha Sperduti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Antoniani
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Fornari
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Brigante
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Giulia Di Rocco
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Simonetta Tagliavini
- Department of Laboratory Medicine and Pathological Anatomy, Azienda Ospedaliero-Universitaria di Modena, NOCSAE, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathological Anatomy, Azienda Ospedaliero-Universitaria di Modena, NOCSAE, Modena, Italy
| | - Daria Morini
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN. Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Modena, Italy
| | - Angela Immacolata Falbo
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN. Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Modena, Italy
| | - Maria Teresa Villani
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN. Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Modena, Italy
| | - Jerzy-Roch Nofer
- Central Laboratory Facility, University Hospital Münster, Münster, Germany
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy; PR China, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Francesco Potì
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
50
|
Yayeh T, Jeong HR, Park YS, Moon S, Sur B, Yoo HS, Oh S. Fumonisin B1-Induced Toxicity Was Not Exacerbated in Glutathione Peroxidase-1/Catalase Double Knock Out Mice. Biomol Ther (Seoul) 2021; 29:52-57. [PMID: 32632050 PMCID: PMC7771844 DOI: 10.4062/biomolther.2020.062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 12/29/2022] Open
Abstract
Fumonisin B1 (FB1) structurally resembles sphingolipids and interferes with their metabolism leading to sphingolipid dysregulation. We questioned if FB1 could exacerbate liver or kidney toxicities in glutathione peroxidase 1 (Gpx1) and catalase (Cat) knockout mice. While higher serum levels of thiobarbituric acid reactive substances (TBARS) and sphinganine (Sa) were measured in Gpx1/Cat knockout mice (Gpx1/Cat KO) than wild type mice after 5 days of FB1 treatment, serum levels of alanine aminotransferase (ALT), sphingosine-1 phosphate (So-1-P), and sphinganine-1 phosphate (Sa-1-P) were found to be relatively low. Although Sa was highly elevated in Gpx1/Cat KO mice and wild mice, lower levels of So and Sa were found in both the kidney and liver tissues of Gpx/Cat KO mice than wild type mice after FB1 treatment. Paradoxically, FB1-induced cellular apoptosis and necrosis were hastened under oxidative stress in Gpx1/Cat KO mice.
Collapse
Affiliation(s)
- Taddesse Yayeh
- Department of Veterinary Science, College of Agriculture and Environmental Sciences, Bahir Dar University, Bahir Dar 5501, Ethiopia
| | - Ha Ram Jeong
- St. Louis College of Pharmacy, St. Louis, MO 63108, USA
| | - Yoon Soo Park
- St. Louis College of Pharmacy, St. Louis, MO 63108, USA
| | - Sohyeon Moon
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Bongjun Sur
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| | - Hwan-Soo Yoo
- College of Pharmacy, Chungbuk National University, Osong 28160, Republic of Korea
| | - Seikwan Oh
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea
| |
Collapse
|