1
|
Lee JO, Lee JM, Kim Y, Park AY, Yoon D, Kim SY, Heo J, Han S, Nam H, Shin HJ, Jeong K, Im M, Kim BJ. Effects of natural killer cell‑conditioned medium on UVB‑induced photoaging in human keratinocytes and a human reconstructed skin model. Mol Med Rep 2025; 31:123. [PMID: 40084708 PMCID: PMC11920773 DOI: 10.3892/mmr.2025.13488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Natural killer (NK) cells produce various cytokines, including interleukin (IL)‑1β, IL‑6, IL‑10, IL‑12, interferon γ, tumor necrosis factor α and transforming growth factor β, which are critical in modulating immune responses. NK cell‑conditioned medium (NK‑CdM), rich in cytokines, has potential applications in therapy and healing. The present study aimed to investigate the protective effect of NK‑CdM against ultraviolet B (UVB)‑mediated photoaging using in vitro and ex vivo models. In human keratinocyte cell line (HaCaT cells), NK‑CdM mitigated UVB‑induced cytotoxicity and suppressed the production of reactive oxygen species. NK‑CdM enhanced the mRNA expression levels of superoxide dismutase 1 (SOD1) and catalase (CAT) and inhibited the reduction in SOD1 and CAT expression levels caused by UVB irradiation. Furthermore, NK‑CdM inhibited the UVB‑mediated nuclear translocation of nuclear factor erythroid 2‑related factor 2. NK‑CdM also prevented UVB‑induced downregulation of filaggrin and involucrin and attenuated the UVB‑induced reduction in hyaluronan synthase (HAS)1, HAS2, HAS3, aquaporin‑3 and hyaluronan levels. Notably, NK‑CdM upregulated the expression of elongation of very long chain fatty acids (ELOVL) enzymes, including ELOVL1, ELOVL5 and ELOVL6, as well as ceramide synthases (CerS), specifically CerS2 and CerS3. Furthermore, NK‑CdM inhibited the UVB‑induced reduction in the levels of these proteins. Overall, these findings suggested that NK‑CdM has the potential to prevent UVB‑mediated photoaging and promote skin health.
Collapse
Affiliation(s)
- Jung Ok Lee
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Jung Min Lee
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Yujin Kim
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - A Yeon Park
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Daewon Yoon
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Su Young Kim
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Jihye Heo
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06974, Republic of Korea
| | - Seungryel Han
- GC Cell Co., Ltd., Yongin, Seoul 16924, Republic of Korea
| | - Hyungjin Nam
- GC Cell Co., Ltd., Yongin, Seoul 16924, Republic of Korea
| | - Hye Jin Shin
- GC Cell Co., Ltd., Yongin, Seoul 16924, Republic of Korea
| | - Kyeongsoo Jeong
- Green Cross Wellbeing Co., Ltd., Yongin, Seoul 16950, Republic of Korea
| | - Minju Im
- Green Cross Wellbeing Co., Ltd., Yongin, Seoul 16950, Republic of Korea
| | - Beom Joon Kim
- Department of Dermatology, College of Medicine, Chung‑Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
2
|
Bamaga AK, Alyazidi AS, Alali FK. A Brief Review of Inherited Neuropathies: A Perspective from Saudi Arabia. Brain Sci 2025; 15:403. [PMID: 40309874 DOI: 10.3390/brainsci15040403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025] Open
Abstract
Inherited neuropathies are a heterogeneous group of disorders that affect the peripheral nervous system, leading to motor, sensory, and autonomic dysfunction. These disorders are classified into various subgroups, including hereditary sensory and motor neuropathies, distal hereditary motor neuropathies, hereditary sensory and autonomic neuropathies, and more complex forms. Advances in genetic testing, particularly next-generation sequencing (NGS), have significantly improved the identification of these disorders. Emerging therapies, such as gene therapy, small molecule therapies, and antisense oligonucleotides, offer promising treatment options. However, current treatments remain limited, and their clinical benefits in humans are not yet fully established. This review provides a comprehensive overview of recent developments and evolving therapeutic options for hereditary neuropathies, focusing on gene therapy, small molecule therapies, and antisense oligonucleotides. It also highlights the current state of inherited neuropathies in Saudi Arabia, emphasizing the need for national guidelines, patient registries, and collaborative research efforts. By integrating advanced genomic technologies and fostering international collaboration, we can improve the diagnosis, management, and treatment outcomes for patients with inherited neuropathies.
Collapse
Affiliation(s)
- Ahmed K Bamaga
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Anas S Alyazidi
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Feryal K Alali
- Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
3
|
Okui M, Noto Y, Kawaguchi J, Iwai N, Wachi M. Biosynthetic Pathways of Alaremycin and Its Derivative: Inhibitors of Porphobilinogen Synthase in Porphyrin Biosynthesis from Streptomyces sp. A012304. ACS BIO & MED CHEM AU 2025; 5:310-319. [PMID: 40255286 PMCID: PMC12006855 DOI: 10.1021/acsbiomedchemau.5c00045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 04/22/2025]
Abstract
The antibiotic alaremycin (5-acetamido-4-oxo-5-hexenoic acid, 1), isolated from Streptomyces sp. A012304, structurally resembles 5-aminolevulinic acid (ALA), a precursor in porphyrin biosynthesis, and inhibits porphobilinogen synthase, the enzyme responsible for catalyzing the first common step of this pathway. In our previous study, the biosynthetic gene cluster responsible for alaremycin production-composed of almA (ALA synthase homologue), almB (N-acetyltransferase), almC (oxidoreductase), and almE (MFS-type transporter)-was identified, and a potential biosynthetic pathway was proposed. In this study, the biosynthetic pathway of 1 was confirmed by detecting intermediates using the liquid chromatography-mass spectrometry/MS (LC-MS/MS) analysis of extracts from Escherichia coli cells transformed with the biosynthetic genes, followed by in vitro reconstitution of the biosynthetic reactions using purified enzymes. AlmA catalyzed the condensation of l-serine and succinyl-CoA to produce 5-amino-6-hydroxy-4-oxohexanoic acid (2), AlmB catalyzed the N-acetylation of 2 to produce 5-acetamido-6-hydroxy-4-oxohexanoic acid (3), and AlmC catalyzed the dehydration of 3 to form 1. The AlmC-catalyzed reaction may involve a two-step mechanism including reduction by NADH and oxidation by Fe3+. Additionally, a novel derivative of 1 was identified in the culture broth of the producer strain, and its structure was determined as 5,6-dihydroalaremycin (5-acetamido-4-oxohexanoic acid, 4). It was revealed that 4 is synthesized via the same biosynthetic pathway but with AlmA and AlmB utilizing l-alanine as the amino acid precursor instead of l-serine.
Collapse
Affiliation(s)
| | | | | | - Noritaka Iwai
- Department of Life Science
and Technology, Institute of Science Tokyo, Yokohama 226-8501, Japan
| | - Masaaki Wachi
- Department of Life Science
and Technology, Institute of Science Tokyo, Yokohama 226-8501, Japan
| |
Collapse
|
4
|
Schäfer JH, Clausmeyer L, Körner C, Esch BM, Wolf VN, Sapia J, Ahmed Y, Walter S, Vanni S, Januliene D, Moeller A, Fröhlich F. Structure of the yeast ceramide synthase. Nat Struct Mol Biol 2025; 32:441-449. [PMID: 39528796 DOI: 10.1038/s41594-024-01415-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 10/01/2024] [Indexed: 11/16/2024]
Abstract
Ceramides are essential lipids involved in forming complex sphingolipids and acting as signaling molecules. They result from the N-acylation of a sphingoid base and a CoA-activated fatty acid, a reaction catalyzed by the ceramide synthase (CerS) family of enzymes. Yet, the precise structural details and catalytic mechanisms of CerSs have remained elusive. Here we used cryo-electron microscopy single-particle analysis to unravel the structure of the yeast CerS complex in both an active and a fumonisin B1-inhibited state. Our results reveal the complex's architecture as a dimer of Lip1 subunits bound to the catalytic subunits Lag1 and Lac1. Each catalytic subunit forms a hydrophobic crevice connecting the cytosolic site with the intermembrane space. The active site, located centrally in the tunnel, was resolved in a substrate preloaded state, representing one intermediate in ceramide synthesis. Our data provide evidence for competitive binding of fumonisin B1 to the acyl-CoA-binding tunnel.
Collapse
Affiliation(s)
- Jan-Hannes Schäfer
- Department of Biology/Chemistry, Structural Biology Section, Osnabrück University, Osnabrück, Germany
| | - Lena Clausmeyer
- Department of Biology/Chemistry, Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany
| | - Carolin Körner
- Department of Biology/Chemistry, Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany
| | - Bianca M Esch
- Department of Biology/Chemistry, Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany
| | - Verena N Wolf
- Department of Biology/Chemistry, Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany
| | - Jennifer Sapia
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Yara Ahmed
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Stefan Walter
- Center of Cellular Nanoanalytic Osnabrück (CellNanOs), Osnabrück University, Osnabrück, Germany
| | - Stefano Vanni
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Swiss National Center for Competence in Research (NCCR), Bio-inspired Materials, University of Fribourg, Fribourg, Switzerland
| | - Dovile Januliene
- Department of Biology/Chemistry, Structural Biology Section, Osnabrück University, Osnabrück, Germany
- Center of Cellular Nanoanalytic Osnabrück (CellNanOs), Osnabrück University, Osnabrück, Germany
| | - Arne Moeller
- Department of Biology/Chemistry, Structural Biology Section, Osnabrück University, Osnabrück, Germany.
- Center of Cellular Nanoanalytic Osnabrück (CellNanOs), Osnabrück University, Osnabrück, Germany.
| | - Florian Fröhlich
- Department of Biology/Chemistry, Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany.
- Center of Cellular Nanoanalytic Osnabrück (CellNanOs), Osnabrück University, Osnabrück, Germany.
| |
Collapse
|
5
|
Murai Y. Elucidation of physiological functions of sphingolipid-related molecules by chemical approaches. Biosci Biotechnol Biochem 2025; 89:205-214. [PMID: 39689917 DOI: 10.1093/bbb/zbae166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 10/22/2024] [Indexed: 12/19/2024]
Abstract
Sphingolipids (SLs), found in all animals, plants, and fungi and in certain prokaryotic organisms, exhibit essential physiological functions that cannot be replicated by other lipids. Although SLs and their related biomolecules behave as lipid mediators, skin barrier systems, and epitopes, their detailed biological functions have not yet been revealed, unlike those of proteins and nucleic acids, because the biosynthesis of SLs is not governed by the central dogma. Recently, SLs have been widely studied in relation to diseases such as obesity, dementia, and neuron agenesis and have attracted attention as molecules related to unmet medical needs. This review presents the recent applications of the SL chemical biology in unmet medical needs.
Collapse
Affiliation(s)
- Yuta Murai
- Division of Applied Bioscience, Graduate School of Agriculture, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
6
|
Imierska M, Zabielski P, Roszczyc-Owsiejczuk K, Pogodzińska K, Błachnio-Zabielska A. Impact of reduced hepatic ceramide levels in high-fat diet mice on glucose metabolism. J Nutr Biochem 2025; 135:109785. [PMID: 39427846 DOI: 10.1016/j.jnutbio.2024.109785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Dysregulation of insulin action in hepatocytes, common in obesity, significantly contributes to insulin resistance, type 2 diabetes, and metabolic syndrome. Previous research highlights ceramides' role in these conditions. This study explores the impact of ceramides by silencing the serine palmitoyltransferase (Sptlc2) gene, crucial for the initial ceramide biosynthesis, using hydrodynamic gene delivery. Male C57BL/6 mice were randomly divided into three groups: one on a low-fat diet (LFD) receiving scrambled shRNA plasmids, another on a high-fat diet (HFD) with scrambled shRNA plasmids, and a third on HFD with a plasmid targeting Sptlc2. Analyses included RT-PCR for gene expression, western blot for protein levels, and UHPLC/MS/MS for lipid profiling. Glucose metabolism was evaluated via oral glucose tolerance tests, homeostatic model assessment of insulin resistance, and glucose-6-phosphate analysis. Results showed that HFD induces insulin resistance by inhibiting insulin signaling and increasing active lipid levels in hepatocytes. Sptlc2 silencing reduced ceramide accumulation, improving insulin signaling and glucose metabolism. Notably, ceramide synthesis inhibition did not significantly affect other lipid levels, highlighting ceramide's critical role in hepatic insulin resistance.
Collapse
Affiliation(s)
- Monika Imierska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Zabielski
- Department of Medical Biology, Medical University of Bialystok, Bialystok, Poland
| | - Kamila Roszczyc-Owsiejczuk
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Karolina Pogodzińska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland
| | - Agnieszka Błachnio-Zabielska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
7
|
Briegas B, Camarero MC, Corbacho J, Labrador J, Sanchez-Vera V, Gavilanes-Ruiz M, Saucedo-García M, Gomez-Jimenez MC. Sphingolipid long chain bases as mediators of cell death in olive fruit abscission. PHYSIOLOGIA PLANTARUM 2025; 177:e70061. [PMID: 39821924 DOI: 10.1111/ppl.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025]
Abstract
Plant sphingolipids are lipophilic membrane components essential for different cellular functions but they also act as signaling molecules in various aspects of plant development. However, the interaction between plant sphingolipids and abscission remains largely uncharacterized. Here, the possible role of sphingolipids in regulating fruit abscission was examined in the abscission zone (AZ) of olive fruit. To this end, sphingolipid levels were manipulated through the application of exogenous sphingolipid long-chain bases (LCBs) or biosynthesis inhibitors, and their effects on fruit abscission as well as sphingolipid LCB/gene expression, hormones, reactive oxygen species (ROS) and cell death levels were examined in the AZ of olive fruit. Our data indicated that exogenous sphinganine (d18:0) induced fruit abscission, whereas the application of sphingosine (d18:1) or phytosphingosine (t18:0) or their phosphorylated derivatives did not have an effect on fruit abscission. Moreover, inhibition of LCB kinase or ceramide synthase, which increases sphingolipid LCB levels in the AZ, reduced fruit break strength. This induction of fruit abscission is associated with elevated ROS levels and cell death in the AZ enriched in salicylic acid (SA) and jasmonic acid (JA). Along the same line, programmed cell death (PCD) was particularly evident on the distal side of the AZ. These data suggest that endogenous d18:0 plays a key cellular role as signaling molecule functioning upstream of the SA/JA signaling pathway in mediating PCD spatially regulated in the AZ during fruit abscission. Overall, the findings reported here provide insight into the complex connection between PCD and plant sphingolipid LCBs, uncovering their interaction in the abscission process.
Collapse
Affiliation(s)
- Beatriz Briegas
- Laboratory of Plant Physiology, Universidad de Extremadura, Badajoz, Spain
| | - Maria C Camarero
- Laboratory of Plant Physiology, Universidad de Extremadura, Badajoz, Spain
| | - Jorge Corbacho
- Laboratory of Plant Physiology, Universidad de Extremadura, Badajoz, Spain
| | - Juana Labrador
- Laboratory of Plant Physiology, Universidad de Extremadura, Badajoz, Spain
| | - Victoria Sanchez-Vera
- Institute for Mediterranean and Subtropical Horticulture La Mayora, University of Malaga-CSIC, Malaga, Spain
| | - Marina Gavilanes-Ruiz
- Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mariana Saucedo-García
- Institute of Agricultural Sciences, Autonomous University of the State of Hidalgo, Tulancingo, Mexico
| | | |
Collapse
|
8
|
Straus AJ, Mavodza G, Senkal CE. Glycosylation of ceramide synthase 6 is required for its activity. J Lipid Res 2025; 66:100715. [PMID: 39608570 PMCID: PMC11732463 DOI: 10.1016/j.jlr.2024.100715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024] Open
Abstract
Sphingolipids play key roles in membrane structure and cellular signaling. Ceramide synthase (CerS)-generated ceramide is implicated in cellular stress responses and induction of apoptosis. Ceramide and other sphingolipids are linked to the induction of ER stress response pathways. However, the mechanisms by which ceramide modulates ER stress signaling are not well understood. Here, we show that the ER stress inducer brefeldin A (BFA) causes increased glycosylation of CerS6, and that treatment with BFA causes increased endogenous ceramide accumulation. To our surprise, we found that CerS6 activity was not affected by BFA-induced glycosylation. Instead, our data show that basal glycosylation of CerS6 at Asn18 is required for CerS6 activity. We used a robust HCT116 CRISPR-Cas9 CerS6 KO with reintroduction of either WT CerS6 or a mutant CerS6 with a point mutation at asparagine-18 to an alanine (N18A) which abrogated glycosylation at that residue. Our data show that cells stably expressing the N18A mutant CerS6 had significantly lower activity in vitro and in situ as compared to WT CerS6 expressing cells. Further, the defective CerS6 with N18A mutation also had defects in GSK3β, AKT, JNK, and STAT3 signaling. Despite being required for CerS6 activity, Asn18 glycosylation did not influence ER stress response pathways. Overall, our study provides vital insight into the regulation of CerS6 activity by posttranslational modification at Asn18 and identifies glycosylation of CerS6 to be important for ceramide generation and regulation of downstream cellular signaling pathways.
Collapse
Affiliation(s)
- Alexandra J Straus
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; C. Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Grace Mavodza
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Can E Senkal
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Massey Comprehensive Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
9
|
Cahoon EB, Kim P, Xie T, González Solis A, Han G, Gong X, Dunn TM. Sphingolipid homeostasis: How do cells know when enough is enough? Implications for plant pathogen responses. PLANT PHYSIOLOGY 2024; 197:kiae460. [PMID: 39222369 DOI: 10.1093/plphys/kiae460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/06/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Sphingolipid homeostatic regulation is important for balancing plant life and death. Plant cells finely tune sphingolipid biosynthesis to ensure sufficient levels to support growth through their basal functions as major components of endomembranes and the plasma membrane. Conversely, accumulation of sphingolipid biosynthetic intermediates, long-chain bases (LCBs) and ceramides, is associated with programmed cell death. Limiting these apoptotic intermediates is important for cell viability, while overriding homeostatic regulation permits cells to generate elevated LCBs and ceramides to respond to pathogens to elicit the hypersensitive response in plant immunity. Key to sphingolipid homeostasis is serine palmitoyltransferase (SPT), an endoplasmic reticulum-associated, multi-subunit enzyme catalyzing the first step in the biosynthesis of LCBs, the defining feature of sphingolipids. Across eukaryotes, SPT interaction with its negative regulator Orosomucoid-like (ORM) is critical for sphingolipid biosynthetic homeostasis. The recent cryo-electron microscopy structure of the Arabidopsis SPT complex indicates that ceramides bind ORMs to competitively inhibit SPT activity. This system provides a sensor for intracellular ceramide concentrations for sphingolipid homeostatic regulation. Combining the newly elucidated Arabidopsis SPT structure and mutant characterization, we present a model for the role of the 2 functionally divergent Arabidopsis ceramide synthase classes to produce ceramides that form repressive (trihydroxy LCB-ceramides) or nonrepressive (dihydroxy LCB-ceramides) ORM interactions to influence SPT activity. We describe how sphingolipid biosynthesis is regulated by the interplay of ceramide synthases with ORM-SPT when "enough is enough" and override homeostatic suppression when "enough is not enough" to respond to environmental stimuli such as microbial pathogen attack.
Collapse
Affiliation(s)
- Edgar B Cahoon
- Center for Plant Science Innovation and Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Panya Kim
- Center for Plant Science Innovation and Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Tian Xie
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ariadna González Solis
- Department of Botany and Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Gongshe Han
- Department of Biochemistry and Molecular Biology, Uniformed Services University of Health Sciences, Bethesda, MD 20814, USA
| | - Xin Gong
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Teresa M Dunn
- Department of Biochemistry and Molecular Biology, Uniformed Services University of Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
10
|
Kim A, Stewart JD. Exploring the Structure-Function Relationships in a 5-Aminolevulinic Acid Synthase and the Use of Protein Engineering to Expand its Substrate Range. Biochemistry 2024. [PMID: 39688068 DOI: 10.1021/acs.biochem.4c00599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
5-Aminolevulinate synthase (ALAS) is a PLP-dependent enzyme that catalyzes the production of 5-aminolevulinate from succinyl-CoA and glycine. Its ability to catalyze the essentially irreversible C-C bond formation has significant potential in chemoenzymatic synthesis of α-amino ketones. Native ALAS, unfortunately, is extremely substrate-selective, and this seriously limits its synthetic utility. Here, we have used three different protein engineering strategies to overcome this problem for the acyl-CoA substrate. By combining previously reported mutation results and structural analysis, a series of site-saturation mutagenesis/screening efforts were focused on R21, T82, N84, and T362 of Rhodopseudomonas palustris ALAS. These yielded single, double, and triple mutants with significantly improved substrate ranges. The steady-state kinetic parameters of several key variants were determined. These data were analyzed in the framework of the ALAS catalytic mechanism to identify the steps that may have been impacted. The most active variant was used in a larger-scale reaction to demonstrate its synthetic potential. Taken together, our results show how ALAS might become a useful biocatalyst for α-amino ketone synthesis and have also allowed us to comment on the relative merits of each the three protein engineering strategies utilized.
Collapse
Affiliation(s)
- Ahram Kim
- Department of Chemistry, University of Florida, 126 Sisler Hall, Gainesville, Florida 32611, United States
| | - Jon D Stewart
- Department of Chemistry, University of Florida, 126 Sisler Hall, Gainesville, Florida 32611, United States
| |
Collapse
|
11
|
Li P, An Z, Sun H, Meng Y, Hou L, Han X, Feng S, Liu Y, Shen S, Zeng F, Dong J, Hao Z. The serine palmitoyltransferase core subunit StLcb2 regulates sphingolipid metabolism and promotes Setosphaeria turcica pathogenicity by modulating appressorium development. Int J Biol Macromol 2024; 283:137928. [PMID: 39579824 DOI: 10.1016/j.ijbiomac.2024.137928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
The fungal pathogen Setosphaeria turcica (S. turcica) causes northern corn leaf blight (NCLB), resulting in significant yield and economic losses in maize. To elucidate the metabolic pathways essential for its pathogenicity, we investigated the metabolome of S. turcica during appressorium development, a critical stage for host infection. Our analysis indicated a substantial enrichment of sphingosine and related compounds during this phase. The application of chemical inhibitors to disrupt sphingolipid metabolism confirmed their pivotal role in appressorium formation and pathogenicity. Additionally, silencing of the serine palmitoyl transferase (Spt) core subunit gene StLCB2 led to significant alterations in fungal morphology and growth, accompanied by changes in cell membrane integrity, surface hydrophobicity, melanin, and sphingosine synthesis. These findings underscore the importance of sphingolipids in the pathogenicity of S. turcica and suggest that targeting specific components of the sphingolipid pathway could aid in developing novel fungicides or genetically engineered maize varieties with increased resistance to NCLB.
Collapse
Affiliation(s)
- Pan Li
- State Key Laboratory of North China Crop Improvement and Regulation/Hebei Bioinformatic Utilization and Technological Innovation Center for Agricultural Microbes, Hebei Agricultural University, Hebei, 071001, China; College of Plant Protection, Hebei Agricultural University, Baoding, Hebei, 071001, China
| | - Zhenwu An
- State Key Laboratory of North China Crop Improvement and Regulation/Hebei Bioinformatic Utilization and Technological Innovation Center for Agricultural Microbes, Hebei Agricultural University, Hebei, 071001, China; College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, 071001, China
| | - Hehe Sun
- College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, 071001, China
| | - Yanan Meng
- College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, 071001, China
| | - Lifeng Hou
- College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, 071001, China
| | - Xinpeng Han
- College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, 071001, China
| | - Shang Feng
- College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, 071001, China
| | - Yuwei Liu
- State Key Laboratory of North China Crop Improvement and Regulation/Hebei Bioinformatic Utilization and Technological Innovation Center for Agricultural Microbes, Hebei Agricultural University, Hebei, 071001, China; College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, 071001, China
| | - Shen Shen
- College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, 071001, China
| | - Fanli Zeng
- State Key Laboratory of North China Crop Improvement and Regulation/Hebei Bioinformatic Utilization and Technological Innovation Center for Agricultural Microbes, Hebei Agricultural University, Hebei, 071001, China; College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, 071001, China.
| | - Jingao Dong
- State Key Laboratory of North China Crop Improvement and Regulation/Hebei Bioinformatic Utilization and Technological Innovation Center for Agricultural Microbes, Hebei Agricultural University, Hebei, 071001, China; College of Plant Protection, Hebei Agricultural University, Baoding, Hebei, 071001, China.
| | - Zhimin Hao
- State Key Laboratory of North China Crop Improvement and Regulation/Hebei Bioinformatic Utilization and Technological Innovation Center for Agricultural Microbes, Hebei Agricultural University, Hebei, 071001, China; College of Life Sciences, Hebei Agricultural University, Baoding, Hebei, 071001, China.
| |
Collapse
|
12
|
de Geus TJ, Franken G, Flinders B, Cuypers E, Joosten EAJ. The Effect of Spinal Cord Stimulation on Spinal Dorsal Horn Lipid Expression in Experimental Painful Diabetic Polyneuropathy: A Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry Imaging Study. Neuromodulation 2024; 27:1360-1371. [PMID: 39425735 DOI: 10.1016/j.neurom.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/07/2024] [Accepted: 09/09/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVES Diabetes-induced peripheral nerve fiber damage can cause painful diabetic polyneuropathy (PDPN), induced by central sensitization through proinflammatory processes in the spinal dorsal horn. Disturbances in spinal dorsal horn lipid metabolism play a major role in proinflammatory regulation. Conventional (Con)-spinal cord stimulation (SCS) is an alternative treatment for pain relief in PDPN, whereas differential target multiplexed (DTM)-SCS could be more effective than Con-SCS, specifically targeting the spinal inflammatory response. We hypothesize that Con- and DTM-SCS differentially affect lipid metabolism in the spinal cord of PDPN animals. To study pain relief mechanisms, we analyzed lipid expression in the spinal dorsal horn using matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry imaging (MSI). MATERIAL AND METHODS Diabetes was induced through streptozotocin (STZ) injection in 28 rats, of which 12 developed PDPN. These and four nondiabetic animals (sham STZ) were implanted with a quadripolar lead and stimulated with Con-, DTM-, or Sham-SCS for 48 hours. Mechanical sensitivity was assessed using Von Frey filaments after 24 and 48 hours. After 48 hours of SCS, the spinal cord was collected, and lipids were analyzed using MALDI-TOF MSI. RESULTS STZ-induced hypersensitivity in the hind paws was reduced by Con- and DTM-SCS. PDPN induction decreased the expression of a glycosphingolipid in laminae 3 of the spinal dorsal horn. After 48 hours of Con- and DTM-SCS, expression levels of several lipids in the spinal dorsal horn decreased, including (HexCer 36:1;O, 40:1;O3), diacylglycerophosphocholines (PC 36:1, 38:6, 40:5), and diacylglycerophosphoserines (PS 36:4). CONCLUSIONS Both Con- and DTM-SCS provide pain relief and decrease spinal dorsal horn lipid expression of PDPN animals, highlighting the complex effects of SCS on the spinal cord physiology. STZ-induced PDPN has a limited effect on lipid expression in the spinal dorsal horn.
Collapse
Affiliation(s)
- Thomas J de Geus
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Research Institute of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.
| | - Glenn Franken
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Research Institute of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Bryn Flinders
- Maastricht MultiModal Molecular Imaging Institute, Division of Mass Spectrometry Imaging, Maastricht University, Maastricht, The Netherlands
| | - Eva Cuypers
- Maastricht MultiModal Molecular Imaging Institute, Division of Mass Spectrometry Imaging, Maastricht University, Maastricht, The Netherlands
| | - Elbert A J Joosten
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Research Institute of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
13
|
Jang Y, Kim CY. The Role of Vitamin E Isoforms and Metabolites in Cancer Prevention: Mechanistic Insights into Sphingolipid Metabolism Modulation. Nutrients 2024; 16:4115. [PMID: 39683509 DOI: 10.3390/nu16234115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Natural forms of vitamin E include four tocopherols and four tocotrienols (α, β, γ, and δ), which are essential as lipophilic antioxidants. Among these eight isoforms, α-tocopherol (αT), the predominant form of vitamin E found in tissues, has traditionally received the most attention in disease prevention research due to its robust antioxidant activity. However, recent studies suggest that other forms of vitamin E exhibit distinct and potentially more potent beneficial activities in disease prevention and treatment. These non-αT forms of vitamin E are metabolized in vivo, producing various metabolites, including 13'-carboxychromanol, though their biological roles remain largely unknown. Notably, sphingolipids, known for their significant roles in cancer biology, may be involved in the anticancer effects of vitamin E through the modulation of sphingolipid metabolism. This review focuses on the diverse biological activities of different vitamin E forms and their metabolites, particularly their anticancer effects, while highlighting the underlying mechanisms, including their novel impact on regulating sphingolipid pathways. By elucidating these interactions, we aim to provide a deeper understanding on the multifaceted roles of vitamin E in cancer prevention and therapy.
Collapse
Affiliation(s)
- Yumi Jang
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 44610, Republic of Korea
- Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Choon Young Kim
- Department of Food and Nutrition, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Human Ecology, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
14
|
Zhang J, Sjøberg KA, Gong S, Wang T, Li F, Kuo A, Durot S, Majcher A, Ardicoglu R, Desgeorges T, Mann CG, Soro Arnáiz I, Fitzgerald G, Gilardoni P, Abel ED, Kon S, Olivares-Villagómez D, Zamboni N, Wolfrum C, Hornemann T, Morscher R, Tisch N, Ghesquière B, Kopf M, Richter EA, De Bock K. Endothelial metabolic control of insulin sensitivity through resident macrophages. Cell Metab 2024; 36:2383-2401.e9. [PMID: 39270655 DOI: 10.1016/j.cmet.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/30/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024]
Abstract
Endothelial cells (ECs) not only form passive blood conduits but actively contribute to nutrient transport and organ homeostasis. The role of ECs in glucose homeostasis is, however, poorly understood. Here, we show that, in skeletal muscle, endothelial glucose transporter 1 (Glut1/Slc2a1) controls glucose uptake via vascular metabolic control of muscle-resident macrophages without affecting transendothelial glucose transport. Lowering endothelial Glut1 via genetic depletion (Glut1ΔEC) or upon a short-term high-fat diet increased angiocrine osteopontin (OPN/Spp1) secretion. This promoted resident muscle macrophage activation and proliferation, which impaired muscle insulin sensitivity. Consequently, co-deleting Spp1 from ECs prevented macrophage accumulation and improved insulin sensitivity in Glut1ΔEC mice. Mechanistically, Glut1-dependent endothelial glucose metabolic rewiring increased OPN in a serine metabolism-dependent fashion. Our data illustrate how the glycolytic endothelium creates a microenvironment that controls resident muscle macrophage phenotype and function and directly links resident muscle macrophages to the maintenance of muscle glucose homeostasis.
Collapse
Affiliation(s)
- Jing Zhang
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Kim Anker Sjøberg
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Songlin Gong
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Tongtong Wang
- Laboratory of Translational Nutritional Biology, Department Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zürich, 8603 Zürich, Switzerland
| | - Fengqi Li
- Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China; Key Laboratory of Immune Response and Immunotherapy, Hefei, China
| | - Andrew Kuo
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Stephan Durot
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Adam Majcher
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland; Institute for Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Raphaela Ardicoglu
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland; Laboratory of Molecular and Behavioral Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Thibaut Desgeorges
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Charlotte Greta Mann
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Ines Soro Arnáiz
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Gillian Fitzgerald
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Paola Gilardoni
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Shigeyuki Kon
- Department of Molecular Immunology, Faculty of Pharmaceutical Sciences, Fukuyama University, Fukuyama, Japan
| | - Danyvid Olivares-Villagómez
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicola Zamboni
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Christian Wolfrum
- Laboratory of Translational Nutritional Biology, Department Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zürich, 8603 Zürich, Switzerland
| | - Thorsten Hornemann
- Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland; Institute for Clinical Chemistry, University Hospital, Zürich, Switzerland
| | - Raphael Morscher
- Pediatric Cancer Metabolism Laboratory, Children`s Research Center, University of Zürich, 8032 Zürich, Switzerland
| | - Nathalie Tisch
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| | - Bart Ghesquière
- Metabolomics Core Facility Leuven, Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Applied Mass Spectrometry, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Manfred Kopf
- Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Erik A Richter
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland.
| |
Collapse
|
15
|
Osinuga A, González Solís A, Cahoon RE, Alsiyabi A, Cahoon EB, Saha R. Deciphering sphingolipid biosynthesis dynamics in Arabidopsis thaliana cell cultures: Quantitative analysis amid data variability. iScience 2024; 27:110675. [PMID: 39297170 PMCID: PMC11409011 DOI: 10.1016/j.isci.2024.110675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/22/2024] [Accepted: 08/01/2024] [Indexed: 09/21/2024] Open
Abstract
Sphingolipids are pivotal for plant development and stress responses. Growing interest has been directed toward fully comprehending the regulatory mechanisms of the sphingolipid pathway. We explore its de novo biosynthesis and homeostasis in Arabidopsis thaliana cell cultures, shedding light on fundamental metabolic mechanisms. Employing 15N isotope labeling and quantitative dynamic modeling approach, we obtained data with notable variations and developed a regularized and constraint-based dynamic metabolic flux analysis (r-DMFA) framework to predict metabolic shifts due to enzymatic changes. Our analysis revealed key enzymes such as sphingoid-base hydroxylase (SBH) and long-chain-base kinase (LCBK) to be critical for maintaining sphingolipid homeostasis. Disruptions in these enzymes were found to affect cellular viability and increase the potential for programmed cell death (PCD). Despite challenges posed by data variability, this work enhances our understanding of sphingolipid metabolism and demonstrates the utility of dynamic modeling in analyzing complex metabolic pathways.
Collapse
Affiliation(s)
- Abraham Osinuga
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Ariadna González Solís
- Department of Biochemistry and Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
- Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Rebecca E Cahoon
- Department of Biochemistry and Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Adil Alsiyabi
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Edgar B Cahoon
- Department of Biochemistry and Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Rajib Saha
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
16
|
Berdyshev E. Skin Lipid Barrier: Structure, Function and Metabolism. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:445-461. [PMID: 39363765 PMCID: PMC11450438 DOI: 10.4168/aair.2024.16.5.445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 10/05/2024]
Abstract
Lipids are important skin components that provide, together with proteins, barrier function of the skin. Keratinocyte terminal differentiation launches unique metabolic changes to lipid metabolism that result in the predominance of ceramides within lipids of the stratum corneum (SC)-the very top portion of the skin. Differentiating keratinocytes form unique ceramides that can be found only in the skin, and generate specialized extracellular structures known as lamellae. Lamellae establish tight hydrophobic layers between dying keratinocytes to protect the body from water loss and also from penetration of allergens and bacteria. Genetic and immunological factors may lead to the failure of keratinocyte terminal differentiation and significantly alter the proportion between SC components. The consequence of such changes is loss or deterioration of skin barrier function that can lead to pathological changes in the skin. This review summarizes our current understanding of the role of lipids in skin barrier function. It also draws attention to the utility of testing SC for lipid and protein biomarkers to predict future onset of allergic skin diseases.
Collapse
Affiliation(s)
- Evgeny Berdyshev
- Department of Medicine, National Jewish Health, Denver, CO, USA.
| |
Collapse
|
17
|
Körner C, Schäfer JH, Esch BM, Parey K, Walter S, Teis D, Januliene D, Schmidt O, Moeller A, Fröhlich F. The structure of the Orm2-containing serine palmitoyltransferase complex reveals distinct inhibitory potentials of yeast Orm proteins. Cell Rep 2024; 43:114627. [PMID: 39167489 DOI: 10.1016/j.celrep.2024.114627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/07/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
Sphingolipid levels are crucial determinants of neurodegenerative disorders and therefore require tight regulation. The Orm protein family and ceramides inhibit the rate-limiting step of sphingolipid biosynthesis-the condensation of L-serine and palmitoyl-coenzyme A (CoA). The yeast isoforms Orm1 and Orm2 form a complex with the serine palmitoyltransferase (SPT). While Orm1 and Orm2 have highly similar sequences, they are differentially regulated, though the mechanistic details remain elusive. Here, we determine the cryoelectron microscopy structure of the SPT complex containing Orm2. Complementary in vitro activity assays and genetic experiments with targeted lipidomics demonstrate a lower activity of the SPT-Orm2 complex than the SPT-Orm1 complex. Our results suggest a higher inhibitory potential of Orm2, despite the similar structures of the Orm1- and Orm2-containing complexes. The high conservation of SPT from yeast to man implies different regulatory capacities for the three human ORMDL isoforms, which might be key for understanding their role in sphingolipid-mediated neurodegenerative disorders.
Collapse
Affiliation(s)
- Carolin Körner
- Bioanalytical Chemistry Section, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
| | - Jan-Hannes Schäfer
- Structural Biology Section, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
| | - Bianca M Esch
- Bioanalytical Chemistry Section, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
| | - Kristian Parey
- Structural Biology Section, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany; Center of Cellular Nanoanalytics Osnabrück (CellNanOs), 49076 Osnabrück, Germany
| | - Stefan Walter
- Center of Cellular Nanoanalytics Osnabrück (CellNanOs), 49076 Osnabrück, Germany
| | - David Teis
- Institute of Molecular Biochemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Dovile Januliene
- Structural Biology Section, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany; Center of Cellular Nanoanalytics Osnabrück (CellNanOs), 49076 Osnabrück, Germany
| | - Oliver Schmidt
- Institute of Cell Biology, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Arne Moeller
- Structural Biology Section, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany; Center of Cellular Nanoanalytics Osnabrück (CellNanOs), 49076 Osnabrück, Germany.
| | - Florian Fröhlich
- Bioanalytical Chemistry Section, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany; Center of Cellular Nanoanalytics Osnabrück (CellNanOs), 49076 Osnabrück, Germany.
| |
Collapse
|
18
|
Šakić Z, Atić A, Potočki S, Bašić-Jukić N. Sphingolipids and Chronic Kidney Disease. J Clin Med 2024; 13:5050. [PMID: 39274263 PMCID: PMC11396415 DOI: 10.3390/jcm13175050] [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: 07/30/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/16/2024] Open
Abstract
Sphingolipids (SLs) are bioactive signaling molecules essential for various cellular processes, including cell survival, proliferation, migration, and apoptosis. Key SLs such as ceramides, sphingosine, and their phosphorylated forms play critical roles in cellular integrity. Dysregulation of SL levels is implicated in numerous diseases, notably chronic kidney disease (CKD). This review focuses on the role of SLs in CKD, highlighting their potential as biomarkers for early detection and prognosis. SLs maintain renal function by modulating the glomerular filtration barrier, primarily through the activity of podocytes. An imbalance in SLs can lead to podocyte damage, contributing to CKD progression. SL metabolism involves complex enzyme-catalyzed pathways, with ceramide serving as a central molecule in de novo and salvage pathways. Ceramides induce apoptosis and are implicated in oxidative stress and inflammation, while sphingosine-1-phosphate (S1P) promotes cell survival and vascular health. Studies have shown that SL metabolism disorders are linked to CKD progression, diabetic kidney disease, and glomerular diseases. Targeting SL pathways could offer novel therapeutic approaches for CKD. This review synthesizes recent research on SL signaling regulation in kidney diseases, emphasizing the importance of maintaining SL balance for renal health and the potential therapeutic benefits of modulating SL pathways.
Collapse
Affiliation(s)
- Zrinka Šakić
- Vuk Vrhovac University Clinic, Dugi dol 4a, 10000 Zagreb, Croatia
| | - Armin Atić
- Division of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, 10000 Zagreb, Croatia
| | - Slavica Potočki
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Nikolina Bašić-Jukić
- Division of Nephrology, Arterial Hypertension, Dialysis and Transplantation, University Hospital Center Zagreb, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
19
|
Jo A, Kim KS, Won J, Shin H, Kim S, Kim B, Kim DJ, Cho JY, Kim HJ. Nasal symbiont Staphylococcus epidermidis restricts influenza A virus replication via the creation of a polyamine-deficient cellular environment. Commun Biol 2024; 7:1031. [PMID: 39174732 PMCID: PMC11341892 DOI: 10.1038/s42003-024-06706-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/08/2024] [Indexed: 08/24/2024] Open
Abstract
Studies on the immune-regulatory roles played by the commensal microbes residing in the nasal mucosa consider the contribution of antiviral immune responses. Here, we sought to identify the nasal microbiome, Staphylococcus epidermidis-regulated antiviral immune responses and the alteration of polyamine metabolites in nasal epithelium. We found that polyamines were required for the life cycle of influenza A virus (IAV) and depletion of polyamines disturbed IAV replication in normal human nasal epithelial (NHNE) cells. Inoculation of S. epidermidis also suppressed IAV infection and the concentration of polyamines including putrescine, spermidine, and spermine was completely attenuated in S. epidermidis-inoculated NHNE cells. S. epidermidis activated the enzyme involved in the production of ornithine from arginine and downregulated the activity of the enzyme involved in the production of putrescine from ornithine in nasal epithelium. S. epidermidis also induced the activation of enzymes that promote the extracellular export of spermine and spermidine in NHNE cells. Our findings demonstrate that S. epidermidis is shown to be able of creating an intracellular environment lacking polyamines in the nasal epithelium and promote the balance of cellular polyamines in favor of the host to restrict influenza virus replication.
Collapse
Affiliation(s)
- Ara Jo
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyeong-Seog Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jina Won
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Haeun Shin
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sujin Kim
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Bora Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Da Jung Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Hyun Jik Kim
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea.
- Seoul National University Hospital, Seoul, Republic of Korea.
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Dingjan T, Futerman AH. Fine-tuned protein-lipid interactions in biological membranes: exploration and implications of the ORMDL-ceramide negative feedback loop in the endoplasmic reticulum. Front Cell Dev Biol 2024; 12:1457209. [PMID: 39170919 PMCID: PMC11335536 DOI: 10.3389/fcell.2024.1457209] [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: 06/30/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Biological membranes consist of a lipid bilayer in which integral membrane proteins are embedded. Based on the compositional complexity of the lipid species found in membranes, and on their specific and selective interactions with membrane proteins, we recently suggested that membrane bilayers can be best described as "finely-tuned molecular machines." We now discuss one such set of lipid-protein interactions by describing a negative feedback mechanism operating in the de novo sphingolipid biosynthetic pathway, which occurs in the membrane of the endoplasmic reticulum, and describe the atomic interactions between the first enzyme in the pathway, namely serine palmitoyl transferase, and the product of the fourth enzyme in the pathway, ceramide. We explore how hydrogen-bonding and hydrophobic interactions formed between Asn13 and Phe63 in the serine palmitoyl transferase complex and ceramide can influence the ceramide content of the endoplasmic reticulum. This example of finely-tuned biochemical interactions raises intriguing mechanistic questions about how sphingolipids and their biosynthetic enzymes could have evolved, particularly in light of their metabolic co-dependence.
Collapse
Affiliation(s)
- Tamir Dingjan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
21
|
Lin L, Lin J, Zhou M, Yuan Y, Li Z. Lipid remodelling and the conversion of lipids into sugars associated with tolerance to cadmium toxicity during white clover seed germination. PHYSIOLOGIA PLANTARUM 2024; 176:e14433. [PMID: 38994561 DOI: 10.1111/ppl.14433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
Cadmium (Cd) is a leading environmental issue worldwide. The current study was conducted to investigate Cd tolerance of 10 commercial white clover (Trifolium repens) cultivars during seed germination and to further explore differences in lipid remodelling, glycometabolism, and the conversion of lipids into sugars contributing to Cd tolerance in the early phase of seedling establishment as well as the accumulation of Cd in seedlings and mature plants. The results show that Cd stress significantly reduced seed germination of 10 cultivars. Compared to Cd-sensitive Sulky, Cd-tolerant Pixie accelerated amylolysis to produce more glucose, fructose, and sucrose by maintaining higher amylase and sucrase activities under Cd stress. Pixie maintained higher contents of various lipids, higher DGDG/MGDG ratio, and lower unsaturation levels of lipids, which could be beneficial to membrane stability and integrity as well as signal transduction in cells after being subjected to Cd stress. In addition, Pixie upregulated expression levels of key genes (TrACX1, TrACX4, TrSDP6, and TrPCK1) involved in the conversion of lipids into sugars for early seedling establishment under Cd stress. These findings indicate that lipid remodelling, enhanced glycometabolism, and accelerated conversion of lipids into sugars are important adaptive strategies for white clover seed germination and subsequent seedling establishment under Cd stress. In addition, Pixie not only accumulated more Cd in seedlings and mature plants than Sulky but also had significantly better growth and phytoremediation efficiency under Cd stress. Pixie could be used as a suitable and critical germplasm for the rehabilitation and re-establishment of Cd-contaminated areas.
Collapse
Affiliation(s)
- Long Lin
- Department of Turf Science and Engineering, College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Junnan Lin
- Department of Turf Science and Engineering, College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Min Zhou
- Department of Turf Science and Engineering, College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yan Yuan
- Department of Turf Science and Engineering, College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Zhou Li
- Department of Turf Science and Engineering, College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
22
|
Ramos-Molina B, Rossell J, Pérez-Montes de Oca A, Pardina E, Genua I, Rojo-López MI, Julián MT, Alonso N, Julve J, Mauricio D. Therapeutic implications for sphingolipid metabolism in metabolic dysfunction-associated steatohepatitis. Front Endocrinol (Lausanne) 2024; 15:1400961. [PMID: 38962680 PMCID: PMC11220194 DOI: 10.3389/fendo.2024.1400961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), a leading cause of chronic liver disease, has increased worldwide along with the epidemics of obesity and related dysmetabolic conditions characterized by impaired glucose metabolism and insulin signaling, such as type 2 diabetes mellitus (T2D). MASLD can be defined as an excessive accumulation of lipid droplets in hepatocytes that occurs when the hepatic lipid metabolism is totally surpassed. This metabolic lipid inflexibility constitutes a central node in the pathogenesis of MASLD and is frequently linked to the overproduction of lipotoxic species, increased cellular stress, and mitochondrial dysfunction. A compelling body of evidence suggests that the accumulation of lipid species derived from sphingolipid metabolism, such as ceramides, contributes significantly to the structural and functional tissue damage observed in more severe grades of MASLD by triggering inflammatory and fibrogenic mechanisms. In this context, MASLD can further progress to metabolic dysfunction-associated steatohepatitis (MASH), which represents the advanced form of MASLD, and hepatic fibrosis. In this review, we discuss the role of sphingolipid species as drivers of MASH and the mechanisms involved in the disease. In addition, given the absence of approved therapies and the limited options for treating MASH, we discuss the feasibility of therapeutic strategies to protect against MASH and other severe manifestations by modulating sphingolipid metabolism.
Collapse
Affiliation(s)
- Bruno Ramos-Molina
- Group of Obesity, Diabetes & Metabolism, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Joana Rossell
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Eva Pardina
- Department de Biochemistry & Molecular Biology, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Idoia Genua
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marina I. Rojo-López
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
| | - María Teresa Julián
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Núria Alonso
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Josep Julve
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Didac Mauricio
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Faculty of Medicine, University of Vic/Central University of Catalonia (UVIC/UCC), Vic, Spain
| |
Collapse
|
23
|
Zhang DK, Song KY, Yan YQ, Zheng JT, Xu J, Da LT, Xu MJ. Structural and mechanistic investigations on CC bond forming α-oxoamine synthase allowing L-glutamate as substrate. Int J Biol Macromol 2024; 268:131696. [PMID: 38642679 DOI: 10.1016/j.ijbiomac.2024.131696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/23/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Carbon‑carbon (C-C) bonds serve as the fundamental structural backbone of organic molecules. As a critical CC bond forming enzyme, α-oxoamine synthase is responsible for the synthesis of α-amino ketones by performing the condensation reaction between amino acids and acyl-CoAs. We previously identified an α-oxoamine synthase (AOS), named as Alb29, involved in albogrisin biosynthesis in Streptomyces albogriseolus MGR072. This enzyme belongs to the α-oxoamine synthase family, a subfamily under the pyridoxal 5'-phosphate (PLP) dependent enzyme superfamily. In this study, we report the crystal structures of Alb29 bound to PLP and L-Glu, which provide the atomic-level structural insights into the substrate recognition by Alb29. We discover that Alb29 can catalyze the amino transformation from L-Gln to L-Glu, besides the condensation of L-Glu with β-methylcrotonyl coenzyme A. Subsequent structural analysis has revealed that one flexible loop in Alb29 plays an important role in both amino transformation and condensation. Based on the crystal structure of the S87G mutant in the loop region, we capture two distinct conformations of the flexible loop in the active site, compared with the wild-type Alb29. Our study offers valuable insights into the catalytic mechanism underlying substrate recognition of Alb29.
Collapse
Affiliation(s)
- Dai-Ke Zhang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Kai-Yuan Song
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Ya-Qian Yan
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Jian-Ting Zheng
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Jun Xu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Lin-Tai Da
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| | - Min-Juan Xu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| |
Collapse
|
24
|
Naruse H, Ishiura H, Esaki K, Mitsui J, Satake W, Greimel P, Shingai N, Machino Y, Kokubo Y, Hamaguchi H, Oda T, Ikkaku T, Yokota I, Takahashi Y, Suzuki Y, Matsukawa T, Goto J, Koh K, Takiyama Y, Morishita S, Yoshikawa T, Tsuji S, Toda T. SPTLC2 variants are associated with early-onset ALS and FTD due to aberrant sphingolipid synthesis. Ann Clin Transl Neurol 2024; 11:946-957. [PMID: 38316966 PMCID: PMC11021611 DOI: 10.1002/acn3.52013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/02/2024] [Accepted: 01/20/2024] [Indexed: 02/07/2024] Open
Abstract
OBJECTIVE Amyotrophic lateral sclerosis (ALS) is a devastating, incurable neurodegenerative disease. A subset of ALS patients manifests with early-onset and complex clinical phenotypes. We aimed to elucidate the genetic basis of these cases to enhance our understanding of disease etiology and facilitate the development of targeted therapies. METHODS Our research commenced with an in-depth genetic and biochemical investigation of two specific families, each with a member diagnosed with early-onset ALS (onset age of <40 years). This involved whole-exome sequencing, trio analysis, protein structure analysis, and sphingolipid measurements. Subsequently, we expanded our analysis to 62 probands with early-onset ALS and further included 440 patients with adult-onset ALS and 1163 healthy controls to assess the prevalence of identified genetic variants. RESULTS We identified heterozygous variants in the serine palmitoyltransferase long chain base subunit 2 (SPTLC2) gene in patients with early-onset ALS. These variants, located in a region closely adjacent to ORMDL3, bear similarities to SPTLC1 variants previously implicated in early-onset ALS. Patients with ALS carrying these SPTLC2 variants displayed elevated plasma ceramide levels, indicative of increased serine palmitoyltransferase (SPT) activity leading to sphingolipid overproduction. INTERPRETATION Our study revealed novel SPTLC2 variants in patients with early-onset ALS exhibiting frontotemporal dementia. The combination of genetic evidence and the observed elevation in plasma ceramide levels establishes a crucial link between dysregulated sphingolipid metabolism and ALS pathogenesis. These findings expand our understanding of ALS's genetic diversity and highlight the distinct roles of gene defects within SPT subunits in its development.
Collapse
Affiliation(s)
- Hiroya Naruse
- Department of Neurology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Precision Medicine Neurology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroyuki Ishiura
- Department of Neurology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of NeurologyOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Kayoko Esaki
- Department of Biotechnology and Life Sciences, Faculty of Biotechnology and Life SciencesSojo UniversityKumamotoJapan
| | - Jun Mitsui
- Department of Neurology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Precision Medicine Neurology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Wataru Satake
- Department of Neurology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Peter Greimel
- Laboratory for Cell Function Dynamics, RIKEN Centre for Brain SciencesWakoSaitamaJapan
| | - Nanoka Shingai
- Division of Applied Life Science, Graduate School of EngineeringSojo UniversityKumamotoJapan
| | - Yuka Machino
- Department of NeurologyNational Hospital Organization Mie National HospitalTsuMieJapan
| | - Yasumasa Kokubo
- Kii ALS/PDC Research Center, Graduate School of Regional Innovation StudiesMie UniversityTsuMieJapan
| | | | - Tetsuya Oda
- Department of NeurologyKita‐Harima Medical CenterOnoHyogoJapan
| | - Tomoko Ikkaku
- Division of NeurologyKobe University Graduate School of MedicineKobeHyogoJapan
- Department of NeurologyHyogo Prefectural Rehabilitation Central HospitalKobeHyogoJapan
| | - Ichiro Yokota
- Division of NeurologyKobe University Graduate School of MedicineKobeHyogoJapan
- Department of NeurologyNational Hospital Organization Hyogo‐Chuo National HospitalSandaHyogoJapan
| | - Yuji Takahashi
- Department of NeurologyNational Center Hospital, National Center of Neurology and PsychiatryTokyoJapan
| | - Yuta Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoChibaJapan
| | - Takashi Matsukawa
- Department of Neurology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Jun Goto
- Department of NeurologyInternational University of Health and Welfare Ichikawa HospitalChibaJapan
| | - Kishin Koh
- Department of Neurology, Graduate School of Medical SciencesUniversity of YamanashiYamanashiJapan
- Department of NeurologyYumura Onsen HospitalYamanashiJapan
| | - Yoshihisa Takiyama
- Department of Neurology, Graduate School of Medical SciencesUniversity of YamanashiYamanashiJapan
- Department of NeurologyFuefuki Central HospitalYamanashiJapan
| | - Shinichi Morishita
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoChibaJapan
| | - Takeo Yoshikawa
- Laboratory of Molecular Psychiatry, RIKEN Center for Brain ScienceWakoSaitamaJapan
| | - Shoji Tsuji
- Department of Neurology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Institute of Medical GenomicsInternational University of Health and WelfareChibaJapan
| | - Tatsushi Toda
- Department of Neurology, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
25
|
Chen Z, Pan Z, Huang C, Zhu X, Li N, Huynh H, Xu J, Huang L, Vaz FM, Liu J, Han Z, Ouyang K. Cardiac lipidomic profiles in mice undergo changes from fetus to adult. Life Sci 2024; 341:122484. [PMID: 38311219 DOI: 10.1016/j.lfs.2024.122484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
AIMS Lipids are essential cellular components with many important biological functions. Disturbed lipid biosynthesis and metabolism has been shown to cause cardiac developmental abnormality and cardiovascular diseases. In this study, we aimed to investigate the composition and the molecular profiles of lipids in mammalian hearts between embryonic and adult stages and uncover the underlying links between lipid and cardiac development and maturation. MATERIALS AND METHODS We collected mouse hearts at the embryonic day 11.5 (E11.5), E15.5, and the age of 2 months, 4 months and 10 months, and performed lipidomic analysis to determine the changes of the composition, molecular species, and relative abundance of cardiac lipids between embryonic and adult stages. Additionally, we also performed the electronic microscopy and RNA sequencing in both embryonic and adult mouse hearts. KEY FINDINGS The relative abundances of certain phospholipids and sphingolipids including cardiolipin, phosphatidylglycerol, phosphatidylethanolamine, and ceramide, are different between embryonic and adult hearts. Such lipidomic changes are accompanied with increased densities of mitochondrial membranes and elevated expression of genes related to mitochondrial formation in adult mouse hearts. We also analyzed individual molecular species of phospholipids and sphingolipids, and revealed that the composition and distribution of lipid molecular species in hearts also change with development. SIGNIFICANCE Our study provides not only a lipidomic view of mammalian hearts when developing from the embryonic to the adult stage, but also a potential pool of lipid indicators for cardiac cell development and maturation.
Collapse
Affiliation(s)
- Ze'e Chen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Zhixiang Pan
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Can Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Xiangbin Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Na Li
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Helen Huynh
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, United States of America
| | - Junjie Xu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam UMC, the Netherlands
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China.
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China.
| |
Collapse
|
26
|
Wang J, Zheng G, Wang L, Meng L, Ren J, Shang L, Li D, Bao Y. Dysregulation of sphingolipid metabolism in pain. Front Pharmacol 2024; 15:1337150. [PMID: 38523645 PMCID: PMC10957601 DOI: 10.3389/fphar.2024.1337150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 02/27/2024] [Indexed: 03/26/2024] Open
Abstract
Pain is a clinical condition that is currently of great concern and is often caused by tissue or nerve damage or occurs as a concomitant symptom of a variety of diseases such as cancer. Severe pain seriously affects the functional status of the body. However, existing pain management programs are not fully satisfactory. Therefore, there is a need to delve deeper into the pathological mechanisms underlying pain generation and to find new targets for drug therapy. Sphingolipids (SLs), as a major component of the bilayer structure of eukaryotic cell membranes, also have powerful signal transduction functions. Sphingolipids are abundant, and their intracellular metabolism constitutes a huge network. Sphingolipids and their various metabolites play significant roles in cell proliferation, differentiation, apoptosis, etc., and have powerful biological activities. The molecules related to sphingolipid metabolism, mainly the core molecule ceramide and the downstream metabolism molecule sphingosine-1-phosphate (S1P), are involved in the specific mechanisms of neurological disorders as well as the onset and progression of various types of pain, and are closely related to a variety of pain-related diseases. Therefore, sphingolipid metabolism can be the focus of research on pain regulation and provide new drug targets and ideas for pain.
Collapse
Affiliation(s)
- Jianfeng Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guangda Zheng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Linfeng Wang
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Linghan Meng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Juanxia Ren
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, China
| | - Lu Shang
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning Province, China
| | - Dongtao Li
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | | |
Collapse
|
27
|
Ikushiro H, Honda T, Murai Y, Murakami T, Takahashi A, Sawai T, Goto H, Ikushiro SI, Miyahara I, Hirabayashi Y, Kamiya N, Monde K, Yano T. Racemization of the substrate and product by serine palmitoyltransferase from Sphingobacterium multivorum yields two enantiomers of the product from d-serine. J Biol Chem 2024; 300:105728. [PMID: 38325740 PMCID: PMC10912632 DOI: 10.1016/j.jbc.2024.105728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
Serine palmitoyltransferase (SPT) catalyzes the pyridoxal-5'-phosphate (PLP)-dependent decarboxylative condensation of l-serine and palmitoyl-CoA to form 3-ketodihydrosphingosine (KDS). Although SPT was shown to synthesize corresponding products from amino acids other than l-serine, it is still arguable whether SPT catalyzes the reaction with d-serine, which is a question of biological importance. Using high substrate and enzyme concentrations, KDS was detected after the incubation of SPT from Sphingobacterium multivorum with d-serine and palmitoyl-CoA. Furthermore, the KDS comprised equal amounts of 2S and 2R isomers. 1H-NMR study showed a slow hydrogen-deuterium exchange at Cα of serine mediated by SPT. We further confirmed that SPT catalyzed the racemization of serine. The rate of the KDS formation from d-serine was comparable to those for the α-hydrogen exchange and the racemization reaction. The structure of the d-serine-soaked crystal (1.65 Å resolution) showed a distinct electron density of the PLP-l-serine aldimine, interpreted as the racemized product trapped in the active site. The structure of the α-methyl-d-serine-soaked crystal (1.70 Å resolution) showed the PLP-α-methyl-d-serine aldimine, mimicking the d-serine-SPT complex prior to racemization. Based on these enzymological and structural analyses, the synthesis of KDS from d-serine was explained as the result of the slow racemization to l-serine, followed by the reaction with palmitoyl-CoA, and SPT would not catalyze the direct condensation between d-serine and palmitoyl-CoA. It was also shown that the S. multivorum SPT catalyzed the racemization of the product KDS, which would explain the presence of (2R)-KDS in the reaction products.
Collapse
Affiliation(s)
- Hiroko Ikushiro
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan.
| | - Takumi Honda
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Yuta Murai
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan; Frontier Research Center for Advanced Material and Life Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido, Japan; Division of Applied Bioscience, Graduate School of Agriculture, Hokkaido University, Sapporo, Hokkaido, Japan.
| | - Taiki Murakami
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka, Japan
| | - Aya Takahashi
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka, Japan
| | - Taiki Sawai
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Haruna Goto
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Shin-Ichi Ikushiro
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, Imizu, Toyama, Japan
| | - Ikuko Miyahara
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, Osaka, Japan
| | - Yoshio Hirabayashi
- RIKEN Cluster for Pioneering Research, RIKEN, Wako, Saitama, Japan; Institute for Environmental and Gender-Specific Medicine, Juntendo University Graduate School of Medicine, Chiba, Japan
| | - Nobuo Kamiya
- Research Center for Artificial Photosynthesis, Osaka Metropolitan University, Osaka, Japan
| | - Kenji Monde
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan; Frontier Research Center for Advanced Material and Life Science, Faculty of Advanced Life Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takato Yano
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Osaka, Japan.
| |
Collapse
|
28
|
Xie T, Dong F, Han G, Wu X, Liu P, Zhang Z, Zhong J, Niranjanakumari S, Gable K, Gupta SD, Liu W, Harrison PJ, Campopiano DJ, Dunn TM, Gong X. Collaborative regulation of yeast SPT-Orm2 complex by phosphorylation and ceramide. Cell Rep 2024; 43:113717. [PMID: 38285738 DOI: 10.1016/j.celrep.2024.113717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/14/2023] [Accepted: 01/12/2024] [Indexed: 01/31/2024] Open
Abstract
The homeostatic regulation of serine palmitoyltransferase (SPT) activity in yeast involves N-terminal phosphorylation of Orm proteins, while higher eukaryotes lack these phosphorylation sites. Although recent studies have indicated a conserved ceramide-mediated feedback inhibition of the SPT-ORM/ORMDL complex in higher eukaryotes, its conservation and relationship with phosphorylation regulation in yeast remain unclear. Here, we determine the structure of the yeast SPT-Orm2 complex in a dephosphomimetic state and identify an evolutionarily conserved ceramide-sensing site. Ceramide stabilizes the dephosphomimetic Orm2 in an inhibitory conformation, facilitated by an intramolecular β-sheet between the N- and C-terminal segments of Orm2. Moreover, we find that a phosphomimetic mutant of Orm2, positioned adjacent to its intramolecular β-sheet, destabilizes the inhibitory conformation of Orm2. Taken together, our findings suggest that both Orm dephosphorylation and ceramide binding are crucial for suppressing SPT activity in yeast. This highlights a distinctive regulatory mechanism in yeast involving the collaborative actions of phosphorylation and ceramide.
Collapse
Affiliation(s)
- Tian Xie
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Feitong Dong
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Gongshe Han
- Department of Biochemistry and Molecular Biology, Uniformed Services University of Health Sciences, Bethesda, MD 20814, USA
| | - Xinyue Wu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Peng Liu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zike Zhang
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jianlong Zhong
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Somashekarappa Niranjanakumari
- Department of Biochemistry and Molecular Biology, Uniformed Services University of Health Sciences, Bethesda, MD 20814, USA
| | - Kenneth Gable
- Department of Biochemistry and Molecular Biology, Uniformed Services University of Health Sciences, Bethesda, MD 20814, USA
| | - Sita D Gupta
- Department of Biochemistry and Molecular Biology, Uniformed Services University of Health Sciences, Bethesda, MD 20814, USA
| | - Wenchen Liu
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Peter J Harrison
- School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK
| | | | - Teresa M Dunn
- Department of Biochemistry and Molecular Biology, Uniformed Services University of Health Sciences, Bethesda, MD 20814, USA.
| | - Xin Gong
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
29
|
Zhou Y, Reynolds TB. Innovations in Antifungal Drug Discovery among Cell Envelope Synthesis Enzymes through Structural Insights. J Fungi (Basel) 2024; 10:171. [PMID: 38535180 PMCID: PMC10970773 DOI: 10.3390/jof10030171] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/16/2024] [Indexed: 11/11/2024] Open
Abstract
Life-threatening systemic fungal infections occur in immunocompromised patients at an alarming rate. Current antifungal therapies face challenges like drug resistance and patient toxicity, emphasizing the need for new treatments. Membrane-bound enzymes account for a large proportion of current and potential antifungal targets, especially ones that contribute to cell wall and cell membrane biosynthesis. Moreover, structural biology has led to a better understanding of the mechanisms by which these enzymes synthesize their products, as well as the mechanism of action for some antifungals. This review summarizes the structures of several current and potential membrane-bound antifungal targets involved in cell wall and cell membrane biosynthesis and their interactions with known inhibitors or drugs. The proposed mechanisms of action for some molecules, gleaned from detailed inhibitor-protein studeis, are also described, which aids in further rational drug design. Furthermore, some potential membrane-bound antifungal targets with known inhibitors that lack solved structures are discussed, as these might be good enzymes for future structure interrogation.
Collapse
Affiliation(s)
| | - Todd B. Reynolds
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA;
| |
Collapse
|
30
|
Ali B, Kumar M, Kumar P, Chauhan A, Usmani SA, Rudramurthy SM, Meis JF, Chakrabarti A, Singh A, Gaur NA, Mondal AK, Prasad R. Sphingolipid diversity in Candida auris: unraveling interclade and drug resistance fingerprints. FEMS Yeast Res 2024; 24:foae008. [PMID: 38444195 PMCID: PMC10941814 DOI: 10.1093/femsyr/foae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/05/2024] [Accepted: 03/04/2024] [Indexed: 03/07/2024] Open
Abstract
In this study, we explored the sphingolipid (SL) landscape in Candida auris, which plays pivotal roles in fungal biology and drug susceptibility. The composition of SLs exhibited substantial variations at both the SL class and molecular species levels among clade isolates. Utilizing principal component analysis, we successfully differentiated the five clades based on their SL class composition. While phytoceramide (PCer) was uniformly the most abundant SL class in all the isolates, other classes showed significant variations. These variations were not limited to SL class level only as the proportion of different molecular species containing variable number of carbons in fatty acid chains also differed between the isolates. Also a comparative analysis revealed abundance of ceramides and glucosylceramides in fluconazole susceptible isolates. Furthermore, by comparing drug-resistant and susceptible isolates within clade IV, we uncovered significant intraclade differences in key SL classes such as high PCer and low long chain base (LCB) content in resistant strains, underscoring the impact of SL heterogeneity on drug resistance development in C. auris. These findings shed light on the multifaceted interplay between genomic diversity, SLs, and drug resistance in this emerging fungal pathogen.
Collapse
Affiliation(s)
- Basharat Ali
- Amity Institute of Integrative Science and Health and Amity Institute of Biotechnology, Amity University Gurgaon, Haryana, 122413, India
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mohit Kumar
- Amity Institute of Integrative Science and Health and Amity Institute of Biotechnology, Amity University Gurgaon, Haryana, 122413, India
- Yeast Biofuel Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067 India
| | - Praveen Kumar
- Amity Institute of Integrative Science and Health and Amity Institute of Biotechnology, Amity University Gurgaon, Haryana, 122413, India
| | - Anshu Chauhan
- Amity Institute of Integrative Science and Health and Amity Institute of Biotechnology, Amity University Gurgaon, Haryana, 122413, India
| | - Sana Akhtar Usmani
- Department of Biochemistry, University of Lucknow, Lucknow, 226007 India
| | | | - Jacques F Meis
- Institute of Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases and Excellence Center for Medical Mycology (ECMM), University of Cologne, Cologne, 50931 Germany
| | | | - Ashutosh Singh
- Department of Biochemistry, University of Lucknow, Lucknow, 226007 India
| | - Naseem A Gaur
- Yeast Biofuel Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067 India
| | - Alok K Mondal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Rajendra Prasad
- Amity Institute of Integrative Science and Health and Amity Institute of Biotechnology, Amity University Gurgaon, Haryana, 122413, India
| |
Collapse
|
31
|
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
|
32
|
Hayashi Y, Tanikawa T. Analysis of the Effects of Brefeldin A on Deuterium-Labeled Sphinganine Metabolism Using Liquid Chromatography-Tandem Mass Spectrometry. Biol Pharm Bull 2024; 47:2173-2181. [PMID: 39756932 DOI: 10.1248/bpb.b24-00292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Ceramide (Cer) is synthesized in the endoplasmic reticulum (ER) using sphinganine as the common backbone and is then transported to the Golgi apparatus to synthesize two complex sphingolipids, sphingomyelin (SM) and glucosylceramide (GlcCer). Brefeldin A (BFA) affects the structure of the Golgi apparatus, resulting in the redistribution of the Golgi proteins into the ER. Therefore, BFA has been used to examine the ER-to-Golgi trafficking of lipids, but the detailed lipid changes in cells upon BFA treatment are not fully understood. Here, we examined the metabolism of deuterium-labeled sphinganine in HEK293T cells treated with BFA using liquid chromatography-tandem mass spectrometry. The BFA-pretreated cells were incubated in the presence of deuterium-labeled sphinganine and BFA for 5 min to 8 h, and the levels of dihydroceramide, Cer, dihydrosphingomyelin, SM, GlcCer, and phosphatidylcholine (PC) were investigated. The levels of Cer species in BFA-treated cells were lower than those in untreated cells within 2 h of incubation, but following >4 h of incubation, the levels of C14-20 Cer, encompassing C14-20 acyl chains, were similar in BFA-treated and untreated cells. Furthermore, BFA treatment increased the levels of C14-20 GlcCer but had little effect on those of C22-24 GlcCer. Moreover, after incubation for >4 h, BFA treatment decreased the levels of saturated C30-32 PC but had almost little effect on those of PC containing a monounsaturated C32-34 acyl chain. Our findings showed that BFA affected the metabolism of various lipids depending on the length and saturation of the fatty acid chain.
Collapse
Affiliation(s)
| | - Takashi Tanikawa
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University
| |
Collapse
|
33
|
Osinuga A, Solis AG, Cahoon RE, Al-Siyabi A, Cahoon EB, Saha R. Quantitative Dynamic Analysis of de novo Sphingolipid Biosynthesis in Arabidopsis thaliana. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.08.570827. [PMID: 38105963 PMCID: PMC10723408 DOI: 10.1101/2023.12.08.570827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Sphingolipids are pivotal for plant development and stress responses. Growing interest has been directed towards fully comprehending the regulatory mechanisms of the sphingolipid pathway. We explore its de novo biosynthesis and homeostasis in Arabidopsis thaliana cell cultures, shedding light on fundamental metabolic mechanisms. Employing 15N isotope labeling and quantitative dynamic modeling approach, we developed a regularized and constraint-based Dynamic Metabolic Flux Analysis (r-DMFA) framework to predict metabolic shifts due to enzymatic changes. Our analysis revealed key enzymes such as sphingoid-base hydroxylase (SBH) and long-chain-base kinase (LCBK) to be critical for maintaining sphingolipid homeostasis. Disruptions in these enzymes were found to affect cellular viability and increase the potential for programmed cell death (PCD). Thus, this work enhances our understanding of sphingolipid metabolism and demonstrates the utility of dynamic modeling in analyzing complex metabolic pathways.
Collapse
Affiliation(s)
- Abraham Osinuga
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Ariadna Gonzalez Solis
- Department of Biochemistry and Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Rebecca E Cahoon
- Department of Biochemistry and Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Adil Al-Siyabi
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Edgar B Cahoon
- Department of Biochemistry and Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Rajib Saha
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
34
|
Darragh IAJ, McNamee N, Daly R, Pacheco SM, O'Driscoll L, Egan B. The separation and identification of circulating small extracellular vesicles from endurance-trained, strength-trained and recreationally active men. J Physiol 2023; 601:5075-5091. [PMID: 37725436 DOI: 10.1113/jp285170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/30/2023] [Indexed: 09/21/2023] Open
Abstract
Small extracellular vesicles (EV) are membrane-encapsulated particles that carry bioactive cargoes, are released by all cell types and are present in all human biofluids. Changes in EV profiles and abundance occur in response to acute exercise, but this study investigated whether individuals with divergent histories of exercise training (recreationally active controls - CON; endurance-trained - END; strength-trained - STR) presented with varied abundances of small EVs in resting samples and whether the abundance of small EVs differed within each group across two measurement days. Participants (n = 38, all male; CON n = 12, END n = 13, STR n = 13) arrived at the lab on two separate occasions in a rested, overnight fasted state, with standardisation of time of day of sampling, recent dietary intake, time since last meal and time since last exercise training session (∼40 h). Whole blood samples were collected and separated into plasma from which small EVs were separated using size exclusion chromatography and identified in accordance with the Minimal Information For Studies of Extracellular Vesicles (MISEV) guidelines. No differences in the abundance of small EVs were observed within or between groups across multiple methods of small EV identification (nanoparticle tracking analysis, flow cytometry, immunoblot of specific EV markers). Targeted metabolomics of the small EV preparations identified 96 metabolites that were associated with the structure and function of small EVs, with no statistically significant differences in concentrations observed across groups. The results of the current study suggest that the abundance and metabolomic profile of small EVs derived from men with divergent histories of exercise training are similar to those in resting blood samples. KEY POINTS: Extracellular vesicles (EV) are membrane-encapsulated particles that are present in circulation and carry bioactive materials as 'cargo'. The abundance and profile of small EVs are responsive to acute exercise, but little is known about the relationship between small EVs and exercise training. This study examined the abundance, and a targeted metabolomic profile, of small EVs separated from the blood of endurance athletes, strength athletes and recreationally active controls at rest (∼40 h after the most recent exercise session) on two separate but identical lab visits. No differences were observed in the abundance or metabolomic profile of small EV preparations between the groups or between the lab visits within each group. Further research should determine whether the bioactive cargoes (e.g. RNA, protein and additional metabolites) carried within EVs are altered in individuals with divergent histories of exercise training or in response to exercise training interventions.
Collapse
Affiliation(s)
- Ian A J Darragh
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Niamh McNamee
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Róisín Daly
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Sarai Martinez Pacheco
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Brendan Egan
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
- Florida Institute for Human and Machine Cognition, Pensacola, Florida, USA
| |
Collapse
|
35
|
Gao T, Li Y, Wang X, Tao R, Ren F. Bifidobacterium longum 68S mediated gut-skin axis homeostasis improved skin barrier damage in aging mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155051. [PMID: 37678055 DOI: 10.1016/j.phymed.2023.155051] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/09/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Bifidobacterium as probiotics, play important roles in skin status, while the potential mechanisms interaction remains unknown. The study further explored the potential mechanism of B. longum 68S in ameliorating skin barrier damage from the perspective of the gut-skin axis in aging mice. METHODS B. longum 68S supplied natural aging mouse model and fecal microbiota transplantation (FMT) experiment proves the key role of intestinal microbiota in B. longum 68S up-regulating the production of ceramide synthesis key enzyme (SPT1) and ceramide level and improving skin barrier damage. Moreover, B. longum 68S supplied SPT1 gene deletion mouse model to investigate the mechanism of B. longum 68S on improving skin barrier damage. RESULTS Transcriptome analysis and 16S rRNA high-throughput pyrosequencing demonstrated that aging mice exhibited skin barrier dysfunction and intestinal dysbiosis. Meanwhile, aging mice exhibited an up-regulation in the trans epidermal water loss (TEWL) and a down-regulation in the level of SPT1, ceramide and skin barrier-related proteins (Loricrin, Keratin 10 and Desmoglein 1). Similarity, the FMT from aging mice to normal mice and SPT1 gene deletion mice could rebuild skin barrier damage and B. longum 68S supplementation exerted a positive effect on it. Further, B. longum 68S-mediated SPT1-derived ceramide production prevented impaired ceramide synthesis-induced endoplasmic reticulum stress and apoptotic response, ultimately improving skin barrier damage in vitro. CONCLUSION Emerging anti-aging therapies are necessary given the poor safety profiles of current pharmaceutical drugs. B. longum 68S may be better alternatives, considering the association between the gut microbiota and healthy aging. The findings suggested that B. longum 68S-mediated gut-skin axis homeostasis, thereby exhibiting an anti-aging effect and facilitate a better understanding of the mechanisms governing the various beneficial effects of B. longum 68S.
Collapse
Affiliation(s)
- Ting Gao
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; Beijing Laboratory of Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; Beijing Laboratory of Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Xiaoyu Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; Beijing Laboratory of Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Ran Tao
- Department of Plastic and Reconstructive Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education; Beijing Laboratory of Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
36
|
Koutsogiannis Z, Mina JG, Albus CA, Kol MA, Holthuis JM, Pohl E, Denny PW. Toxoplasma ceramide synthases: Gene duplication, functional divergence, and roles in parasite fitness. FASEB J 2023; 37:e23229. [PMID: 37795915 PMCID: PMC10946778 DOI: 10.1096/fj.202201603rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 08/16/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023]
Abstract
Toxoplasma gondii is an obligate, intracellular apicomplexan protozoan parasite of both humans and animals that can cause fetal damage and abortion and severe disease in the immunosuppressed. Sphingolipids have indispensable functions as signaling molecules and are essential and ubiquitous components of eukaryotic membranes that are both synthesized and scavenged by the Apicomplexa. Ceramide is the precursor for all sphingolipids, and here we report the identification, localization and analyses of the Toxoplasma ceramide synthases TgCerS1 and TgCerS2. Interestingly, we observed that while TgCerS1 was a fully functional orthologue of the yeast ceramide synthase (Lag1p) capable of catalyzing the conversion of sphinganine to ceramide, in contrast TgCerS2 was catalytically inactive. Furthermore, genomic deletion of TgCerS1 using CRISPR/Cas-9 led to viable but slow-growing parasites indicating its importance but not indispensability. In contrast, genomic knock out of TgCerS2 was only accessible utilizing the rapamycin-inducible Cre recombinase system. Surprisingly, the results demonstrated that this "pseudo" ceramide synthase, TgCerS2, has a considerably greater role in parasite fitness than its catalytically active orthologue (TgCerS1). Phylogenetic analyses indicated that, as in humans and plants, the ceramide synthase isoforms found in Toxoplasma and other Apicomplexa may have arisen through gene duplication. However, in the Apicomplexa the duplicated copy is hypothesized to have subsequently evolved into a non-functional "pseudo" ceramide synthase. This arrangement is unique to the Apicomplexa and further illustrates the unusual biology that characterize these protozoan parasites.
Collapse
Affiliation(s)
| | - John G. Mina
- Department of BiosciencesDurham UniversityDurhamUK
| | | | - Matthijs A. Kol
- Molecular Cell Biology Division, Department of Biology/ChemistryUniversity of OsnabrückOsnabrückGermany
| | - Joost C. M. Holthuis
- Molecular Cell Biology Division, Department of Biology/ChemistryUniversity of OsnabrückOsnabrückGermany
| | - Ehmke Pohl
- Department of BiosciencesDurham UniversityDurhamUK
- Department of ChemistryDurham UniversityDurhamUK
| | | |
Collapse
|
37
|
Schäfer JH, Körner C, Esch BM, Limar S, Parey K, Walter S, Januliene D, Moeller A, Fröhlich F. Structure of the ceramide-bound SPOTS complex. Nat Commun 2023; 14:6196. [PMID: 37794019 PMCID: PMC10550967 DOI: 10.1038/s41467-023-41747-z] [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] [Received: 02/03/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023] Open
Abstract
Sphingolipids are structural membrane components that also function in cellular stress responses. The serine palmitoyltransferase (SPT) catalyzes the rate-limiting step in sphingolipid biogenesis. Its activity is tightly regulated through multiple binding partners, including Tsc3, Orm proteins, ceramides, and the phosphatidylinositol-4-phosphate (PI4P) phosphatase Sac1. The structural organization and regulatory mechanisms of this complex are not yet understood. Here, we report the high-resolution cryo-EM structures of the yeast SPT in complex with Tsc3 and Orm1 (SPOT) as dimers and monomers and a monomeric complex further carrying Sac1 (SPOTS). In all complexes, the tight interaction of the downstream metabolite ceramide and Orm1 reveals the ceramide-dependent inhibition. Additionally, observation of ceramide and ergosterol binding suggests a co-regulation of sphingolipid biogenesis and sterol metabolism within the SPOTS complex.
Collapse
Affiliation(s)
- Jan-Hannes Schäfer
- Osnabrück University Department of Biology/Chemistry Structural Biology section, 49076, Osnabrück, Germany
| | - Carolin Körner
- Osnabrück University Department of Biology/Chemistry Bioanalytical Chemistry section, 49076, Osnabrück, Germany
| | - Bianca M Esch
- Osnabrück University Department of Biology/Chemistry Bioanalytical Chemistry section, 49076, Osnabrück, Germany
| | - Sergej Limar
- Osnabrück University Department of Biology/Chemistry Bioanalytical Chemistry section, 49076, Osnabrück, Germany
| | - Kristian Parey
- Osnabrück University Department of Biology/Chemistry Structural Biology section, 49076, Osnabrück, Germany
- Osnabrück University Center of Cellular Nanoanalytic Osnabrück (CellNanOs), 49076, Osnabrück, Germany
| | - Stefan Walter
- Osnabrück University Center of Cellular Nanoanalytic Osnabrück (CellNanOs), 49076, Osnabrück, Germany
| | - Dovile Januliene
- Osnabrück University Department of Biology/Chemistry Structural Biology section, 49076, Osnabrück, Germany.
- Osnabrück University Center of Cellular Nanoanalytic Osnabrück (CellNanOs), 49076, Osnabrück, Germany.
| | - Arne Moeller
- Osnabrück University Department of Biology/Chemistry Structural Biology section, 49076, Osnabrück, Germany.
- Osnabrück University Center of Cellular Nanoanalytic Osnabrück (CellNanOs), 49076, Osnabrück, Germany.
| | - Florian Fröhlich
- Osnabrück University Department of Biology/Chemistry Bioanalytical Chemistry section, 49076, Osnabrück, Germany.
- Osnabrück University Center of Cellular Nanoanalytic Osnabrück (CellNanOs), 49076, Osnabrück, Germany.
| |
Collapse
|
38
|
Mattioli R, Ilari A, Colotti B, Mosca L, Fazi F, Colotti G. Doxorubicin and other anthracyclines in cancers: Activity, chemoresistance and its overcoming. Mol Aspects Med 2023; 93:101205. [PMID: 37515939 DOI: 10.1016/j.mam.2023.101205] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023]
Abstract
Anthracyclines have been important and effective treatments against a number of cancers since their discovery. However, their use in therapy has been complicated by severe side effects and toxicity that occur during or after treatment, including cardiotoxicity. The mode of action of anthracyclines is complex, with several mechanisms proposed. It is possible that their high toxicity is due to the large set of processes involved in anthracycline action. The development of resistance is a major barrier to successful treatment when using anthracyclines. This resistance is based on a series of mechanisms that have been studied and addressed in recent years. This work provides an overview of the anthracyclines used in cancer therapy. It discusses their mechanisms of activity, toxicity, and chemoresistance, as well as the approaches used to improve their activity, decrease their toxicity, and overcome resistance.
Collapse
Affiliation(s)
- Roberto Mattioli
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Andrea Ilari
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy
| | - Beatrice Colotti
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Luciana Mosca
- Dept. Biochemical Sciences A. Rossi Fanelli, Sapienza University of Rome, Rome, Italy
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council IBPM-CNR, Rome, Italy.
| |
Collapse
|
39
|
Ogonkov A, Dieterich CL, Meoded RA, Piel J, Fraley AE, Sasso S. Characterization of an Unusual α-Oxoamine Synthase Off-Loading Domain from a Cyanobacterial Type I Fatty Acid Synthase. Chembiochem 2023; 24:e202300209. [PMID: 37144248 DOI: 10.1002/cbic.202300209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/06/2023]
Abstract
Type I fatty acid synthases (FASs) are known from higher eukaryotes and fungi. We report the discovery of FasT, a rare type I FAS from the cyanobacterium Chlorogloea sp. CCALA695. FasT possesses an unusual off-loading domain, which was heterologously expressed in E. coli and found to act as an α-oxoamine synthase (AOS) in vitro. Similar to serine palmitoyltransferases from sphingolipid biosynthesis, the AOS off-loading domain catalyzes a decarboxylative Claisen condensation between l-serine and a fatty acyl thioester. While the AOS domain was strictly specific for l-serine, thioesters with saturated fatty acyl chains of six carbon atoms and longer were tolerated, with the highest activity observed for stearoyl-coenzyme A (C18 ). Our findings suggest a novel route to α-amino ketones via the direct condensation of iteratively produced long-chain fatty acids with l-serine by a FAS with a cis-acting AOS off-loading domain.
Collapse
Affiliation(s)
- Andrei Ogonkov
- Department of Biology, Institute of Microbiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
- Institute of Biology, Leipzig University, Johannisallee 23, 04107, Leipzig, Germany
| | - Cora L Dieterich
- Department of Biology, Institute of Microbiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Roy A Meoded
- Department of Biology, Institute of Microbiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Jörn Piel
- Department of Biology, Institute of Microbiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Amy E Fraley
- Department of Biology, Institute of Microbiology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093, Zurich, Switzerland
| | - Severin Sasso
- Institute of Biology, Leipzig University, Johannisallee 23, 04107, Leipzig, Germany
| |
Collapse
|
40
|
Issleny BM, Jamjoum R, Majumder S, Stiban J. Sphingolipids: From structural components to signaling hubs. Enzymes 2023; 54:171-201. [PMID: 37945171 DOI: 10.1016/bs.enz.2023.07.003] [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] [Indexed: 11/12/2023]
Abstract
In late November 2019, Prof. Lina M. Obeid passed away from cancer, a disease she spent her life researching and studying its intricate molecular underpinnings. Along with her husband, Prof. Yusuf A. Hannun, Obeid laid down the foundations of sphingolipid biochemistry and oversaw its remarkable evolution over the years. Lipids are a class of macromolecules that are primarily associated with cellular architecture. In fact, lipids constitute the perimeter of the cell in such a way that without them, there cannot be cells. Hence, much of the early research on lipids identified the function of this class of biological molecules as merely structural. Nevertheless, unlike proteins, carbohydrates, and nucleic acids, lipids are elaborately diverse as they are not made up of monomers in polymeric forms. This diversity in structure is clearly mirrored by functional pleiotropy. In this chapter, we focus on a major subset of lipids, sphingolipids, and explore their historic rise from merely inert structural components of plasma membranes to lively and necessary signaling molecules that transmit various signals and control many cellular processes. We will emphasize the works of Lina Obeid since she was an integral pillar of the sphingolipid research world.
Collapse
Affiliation(s)
- Batoul M Issleny
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | - Rama Jamjoum
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | | | - Johnny Stiban
- Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine.
| |
Collapse
|
41
|
Wang W, Sherry T, Cheng X, Fan Q, Cornell R, Liu J, Xiao Z, Pocock R. An intestinal sphingolipid confers intergenerational neuroprotection. Nat Cell Biol 2023; 25:1196-1207. [PMID: 37537365 PMCID: PMC10415181 DOI: 10.1038/s41556-023-01195-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 06/27/2023] [Indexed: 08/05/2023]
Abstract
In animals, maternal diet and environment can influence the health of offspring. Whether and how maternal dietary choice impacts the nervous system across multiple generations is not well understood. Here we show that feeding Caenorhabditis elegans with ursolic acid, a natural plant product, improves axon transport and reduces adult-onset axon fragility intergenerationally. Ursolic acid provides neuroprotection by enhancing maternal provisioning of sphingosine-1-phosphate, a bioactive sphingolipid. Intestine-to-oocyte sphingosine-1-phosphate transfer is required for intergenerational neuroprotection and is dependent on the RME-2 lipoprotein yolk receptor. Sphingosine-1-phosphate acts intergenerationally by upregulating the transcription of the acid ceramidase-1 (asah-1) gene in the intestine. Spatial regulation of sphingolipid metabolism is critical, as inappropriate asah-1 expression in neurons causes developmental axon outgrowth defects. Our results show that sphingolipid homeostasis impacts the development and intergenerational health of the nervous system. The ability of specific lipid metabolites to act as messengers between generations may have broad implications for dietary choice during reproduction.
Collapse
Affiliation(s)
- Wenyue Wang
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Tessa Sherry
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Xinran Cheng
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Qi Fan
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Rebecca Cornell
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Jie Liu
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Zhicheng Xiao
- Neuroscience Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
42
|
Najjar RS. The Impacts of Animal-Based Diets in Cardiovascular Disease Development: A Cellular and Physiological Overview. J Cardiovasc Dev Dis 2023; 10:282. [PMID: 37504538 PMCID: PMC10380617 DOI: 10.3390/jcdd10070282] [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: 05/24/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the United States, and diet plays an instrumental role in CVD development. Plant-based diets have been strongly tied to a reduction in CVD incidence. In contrast, animal food consumption may increase CVD risk. While increased serum low-density lipoprotein (LDL) cholesterol concentrations are an established risk factor which may partially explain the positive association with animal foods and CVD, numerous other biochemical factors are also at play. Thus, the aim of this review is to summarize the major cellular and molecular effects of animal food consumption in relation to CVD development. Animal-food-centered diets may (1) increase cardiovascular toll-like receptor (TLR) signaling, due to increased serum endotoxins and oxidized LDL cholesterol, (2) increase cardiovascular lipotoxicity, (3) increase renin-angiotensin system components and subsequent angiotensin II type-1 receptor (AT1R) signaling and (4) increase serum trimethylamine-N-oxide concentrations. These nutritionally mediated factors independently increase cardiovascular oxidative stress and inflammation and are all independently tied to CVD development. Public policy efforts should continue to advocate for the consumption of a mostly plant-based diet, with the minimization of animal-based foods.
Collapse
Affiliation(s)
- Rami Salim Najjar
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
43
|
Taylor CJ, Felton KC, Wigh D, Jeraal MI, Grainger R, Chessari G, Johnson CN, Lapkin AA. Accelerated Chemical Reaction Optimization Using Multi-Task Learning. ACS CENTRAL SCIENCE 2023; 9:957-968. [PMID: 37252348 PMCID: PMC10214532 DOI: 10.1021/acscentsci.3c00050] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Indexed: 05/31/2023]
Abstract
Functionalization of C-H bonds is a key challenge in medicinal chemistry, particularly for fragment-based drug discovery (FBDD) where such transformations require execution in the presence of polar functionality necessary for protein binding. Recent work has shown the effectiveness of Bayesian optimization (BO) for the self-optimization of chemical reactions; however, in all previous cases these algorithmic procedures have started with no prior information about the reaction of interest. In this work, we explore the use of multitask Bayesian optimization (MTBO) in several in silico case studies by leveraging reaction data collected from historical optimization campaigns to accelerate the optimization of new reactions. This methodology was then translated to real-world, medicinal chemistry applications in the yield optimization of several pharmaceutical intermediates using an autonomous flow-based reactor platform. The use of the MTBO algorithm was shown to be successful in determining optimal conditions of unseen experimental C-H activation reactions with differing substrates, demonstrating an efficient optimization strategy with large potential cost reductions when compared to industry-standard process optimization techniques. Our findings highlight the effectiveness of the methodology as an enabling tool in medicinal chemistry workflows, representing a step-change in the utilization of data and machine learning with the goal of accelerated reaction optimization.
Collapse
Affiliation(s)
- Connor J. Taylor
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
- Innovation
Centre in Digital Molecular Technologies, Yusuf Hamied Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, United
Kingdom
| | - Kobi C. Felton
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United Kingdom
| | - Daniel Wigh
- Innovation
Centre in Digital Molecular Technologies, Yusuf Hamied Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, United
Kingdom
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United Kingdom
| | - Mohammed I. Jeraal
- Cambridge
Centre for Advanced Research and Education in Singapore Ltd., 1 Create Way, CREATE Tower #05-05, 138602, Singapore
| | - Rachel Grainger
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Gianni Chessari
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Christopher N. Johnson
- Astex
Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge, CB4 0QA, United Kingdom
| | - Alexei A. Lapkin
- Innovation
Centre in Digital Molecular Technologies, Yusuf Hamied Department
of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, United
Kingdom
- Department
of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, Cambridge CB3 0AS, United Kingdom
- Cambridge
Centre for Advanced Research and Education in Singapore Ltd., 1 Create Way, CREATE Tower #05-05, 138602, Singapore
| |
Collapse
|
44
|
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
|
45
|
Srivastava S, Shaked HM, Gable K, Gupta SD, Pan X, Somashekarappa N, Han G, Mohassel P, Gotkine M, Doney E, Goldenberg P, Tan QKG, Gong Y, Kleinstiver B, Wishart B, Cope H, Pires CB, Stutzman H, Spillmann RC, Sadjadi R, Elpeleg O, Lee CH, Bellen HJ, Edvardson S, Eichler F, Dunn TM, Dai H, Dhar SU, Emrick LT, Goldman AM, Hanchard NA, Jamal F, Karaviti L, Lalani SR, Lee BH, Lewis RA, Marom R, Moretti PM, Murdock DR, Nicholas SK, Orengo JP, Posey JE, Potocki L, Rosenfeld JA, Samson SL, Scott DA, Tran AA, Vogel TP, Wangler MF, Yamamoto S, Eng CM, Liu P, Ward PA, Behrens E, Deardorff M, Falk M, Hassey K, Sullivan K, Vanderver A, Goldstein DB, Cope H, McConkie-Rosell A, Schoch K, Shashi V, Smith EC, Spillmann RC, Sullivan JA, Tan QKG, Walley NM, Agrawal PB, Beggs AH, Berry GT, Briere LC, Cobban LA, Coggins M, Cooper CM, Fieg EL, High F, Holm IA, Korrick S, Krier JB, Lincoln SA, Loscalzo J, Maas RL, MacRae CA, Pallais JC, Rao DA, Rodan LH, Silverman EK, Stoler JM, Sweetser DA, Walker M, Walsh CA, Esteves C, Kelley EG, Kohane IS, LeBlanc K, McCray AT, Nagy A, Dasari S, et alSrivastava S, Shaked HM, Gable K, Gupta SD, Pan X, Somashekarappa N, Han G, Mohassel P, Gotkine M, Doney E, Goldenberg P, Tan QKG, Gong Y, Kleinstiver B, Wishart B, Cope H, Pires CB, Stutzman H, Spillmann RC, Sadjadi R, Elpeleg O, Lee CH, Bellen HJ, Edvardson S, Eichler F, Dunn TM, Dai H, Dhar SU, Emrick LT, Goldman AM, Hanchard NA, Jamal F, Karaviti L, Lalani SR, Lee BH, Lewis RA, Marom R, Moretti PM, Murdock DR, Nicholas SK, Orengo JP, Posey JE, Potocki L, Rosenfeld JA, Samson SL, Scott DA, Tran AA, Vogel TP, Wangler MF, Yamamoto S, Eng CM, Liu P, Ward PA, Behrens E, Deardorff M, Falk M, Hassey K, Sullivan K, Vanderver A, Goldstein DB, Cope H, McConkie-Rosell A, Schoch K, Shashi V, Smith EC, Spillmann RC, Sullivan JA, Tan QKG, Walley NM, Agrawal PB, Beggs AH, Berry GT, Briere LC, Cobban LA, Coggins M, Cooper CM, Fieg EL, High F, Holm IA, Korrick S, Krier JB, Lincoln SA, Loscalzo J, Maas RL, MacRae CA, Pallais JC, Rao DA, Rodan LH, Silverman EK, Stoler JM, Sweetser DA, Walker M, Walsh CA, Esteves C, Kelley EG, Kohane IS, LeBlanc K, McCray AT, Nagy A, Dasari S, Lanpher BC, Lanza IR, Morava E, Oglesbee D, Bademci G, Barbouth D, Bivona S, Carrasquillo O, Chang TCP, Forghani I, Grajewski A, Isasi R, Lam B, Levitt R, Liu XZ, McCauley J, Sacco R, Saporta M, Schaechter J, Tekin M, Telischi F, Thorson W, Zuchner S, Colley HA, Dayal JG, Eckstein DJ, Findley LC, Krasnewich DM, Mamounas LA, Manolio TA, Mulvihill JJ, LaMoure GL, Goldrich MP, Urv TK, Doss AL, Acosta MT, Bonnenmann C, D’Souza P, Draper DD, Ferreira C, Godfrey RA, Groden CA, Macnamara EF, Maduro VV, Markello TC, Nath A, Novacic D, Pusey BN, Toro C, Wahl CE, Baker E, Burke EA, Adams DR, Gahl WA, Malicdan MCV, Tifft CJ, Wolfe LA, Yang J, Power B, Gochuico B, Huryn L, Latham L, Davis J, Mosbrook-Davis D, Rossignol F, Solomon B, MacDowall J, Thurm A, Zein W, Yousef M, Adam M, Amendola L, Bamshad M, Beck A, Bennett J, Berg-Rood B, Blue E, Boyd B, Byers P, Chanprasert S, Cunningham M, Dipple K, Doherty D, Earl D, Glass I, Golden-Grant K, Hahn S, Hing A, Hisama FM, Horike-Pyne M, Jarvik GP, Jarvik J, Jayadev S, Lam C, Maravilla K, Mefford H, Merritt JL, Mirzaa G, Nickerson D, Raskind W, Rosenwasser N, Scott CR, Sun A, Sybert V, Wallace S, Wener M, Wenger T, Ashley EA, Bejerano G, Bernstein JA, Bonner D, Coakley TR, Fernandez L, Fisher PG, Fresard L, Hom J, Huang Y, Kohler JN, Kravets E, Majcherska MM, Martin BA, Marwaha S, McCormack CE, Raja AN, Reuter CM, Ruzhnikov M, Sampson JB, Smith KS, Sutton S, Tabor HK, Tucker BM, Wheeler MT, Zastrow DB, Zhao C, Byrd WE, Crouse AB, Might M, Nakano-Okuno M, Whitlock J, Brown G, Butte MJ, Dell’Angelica EC, Dorrani N, Douine ED, Fogel BL, Gutierrez I, Huang A, Krakow D, Lee H, Loo SK, Mak BC, Martin MG, Martínez-Agosto JA, McGee E, Nelson SF, Nieves-Rodriguez S, Palmer CGS, Papp JC, Parker NH, Renteria G, Signer RH, Sinsheimer JS, Wan J, Wang LK, Perry KW, Woods JD, Alvey J, Andrews A, Bale J, Bohnsack J, Botto L, Carey J, Pace L, Longo N, Marth G, Moretti P, Quinlan A, Velinder M, Viskochi D, Bayrak-Toydemir P, Mao R, Westerfield M, Bican A, Brokamp E, Duncan L, Hamid R, Kennedy J, Kozuira M, Newman JH, PhillipsIII JA, Rives L, Robertson AK, Solem E, Cogan JD, Cole FS, Hayes N, Kiley D, Sisco K, Wambach J, Wegner D, Baldridge D, Pak S, Schedl T, Shin J, Solnica-Krezel L, Sadjadi R, Elpeleg O, Lee CH, Bellen HJ, Edvardson S, Eichler F, Dunn TM. SPTSSA variants alter sphingolipid synthesis and cause a complex hereditary spastic paraplegia. Brain 2023; 146:1420-1435. [PMID: 36718090 PMCID: PMC10319774 DOI: 10.1093/brain/awac460] [Show More Authors] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 11/03/2022] [Accepted: 11/19/2022] [Indexed: 02/01/2023] Open
Abstract
Sphingolipids are a diverse family of lipids with critical structural and signalling functions in the mammalian nervous system, where they are abundant in myelin membranes. Serine palmitoyltransferase, the enzyme that catalyses the rate-limiting reaction of sphingolipid synthesis, is composed of multiple subunits including an activating subunit, SPTSSA. Sphingolipids are both essential and cytotoxic and their synthesis must therefore be tightly regulated. Key to the homeostatic regulation are the ORMDL proteins that are bound to serine palmitoyltransferase and mediate feedback inhibition of enzymatic activity when sphingolipid levels become excessive. Exome sequencing identified potential disease-causing variants in SPTSSA in three children presenting with a complex form of hereditary spastic paraplegia. The effect of these variants on the catalytic activity and homeostatic regulation of serine palmitoyltransferase was investigated in human embryonic kidney cells, patient fibroblasts and Drosophila. Our results showed that two different pathogenic variants in SPTSSA caused a hereditary spastic paraplegia resulting in progressive motor disturbance with variable sensorineural hearing loss and language/cognitive dysfunction in three individuals. The variants in SPTSSA impaired the negative regulation of serine palmitoyltransferase by ORMDLs leading to excessive sphingolipid synthesis based on biochemical studies and in vivo studies in Drosophila. These findings support the pathogenicity of the SPTSSA variants and point to excessive sphingolipid synthesis due to impaired homeostatic regulation of serine palmitoyltransferase as responsible for defects in early brain development and function.
Collapse
Affiliation(s)
- Siddharth Srivastava
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, BostonChildren's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hagar Mor Shaked
- Department of Genetics, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Kenneth Gable
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sita D Gupta
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Xueyang Pan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Niranjanakumari Somashekarappa
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Gongshe Han
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Payam Mohassel
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20814, USA
| | - Marc Gotkine
- Department of Genetics, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | | | - Paula Goldenberg
- Department of Pediatrics, Section on Medical Genetics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Queenie K G Tan
- Department of Pediatrics, Division of Medical Genetics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yi Gong
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Benjamin Kleinstiver
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.,Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA.,Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Brian Wishart
- Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Heidi Cope
- Department of Pediatrics, Division of Medical Genetics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Claudia Brito Pires
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Hannah Stutzman
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.,Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Rebecca C Spillmann
- Department of Pediatrics, Division of Medical Genetics, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Reza Sadjadi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Orly Elpeleg
- Department of Genetics, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Chia-Hsueh Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Simon Edvardson
- Pediatric Neurology Unit, Hadassah University Hospital, Mount Scopus, Jerusalem 91240, Israel
| | - Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.,Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Teresa M Dunn
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Reza Sadjadi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School , Boston, MA 02114 , USA
| | - Orly Elpeleg
- Department of Genetics, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem , Jerusalem 91120 , Israel
| | - Chia-Hsueh Lee
- Department of Structural Biology, St. Jude Children’s Research Hospital , Memphis, TN 38105 , USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine , Houston, TX 77030 , USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital , Houston, TX 77030 , USA
| | - Simon Edvardson
- Pediatric Neurology Unit, Hadassah University Hospital, Mount Scopus , Jerusalem 91240 , Israel
| | - Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School , Boston, MA 02114 , USA
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School , Boston, MA 02114 , USA
| | - Teresa M Dunn
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences , Bethesda, MD 20814 , USA
| | | |
Collapse
|
46
|
Julca I, Vargas P, Gabaldón T. Phylogenomics of the Olea europaea complex using 15 whole genomes supports recurrent genetic admixture together with differentiation into seven subspecies. BMC Biol 2023; 21:85. [PMID: 37069619 PMCID: PMC10111821 DOI: 10.1186/s12915-023-01583-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND The last taxonomic account of Olea recognises six subspecies within Olea europaea L., including the Mediterranean olive tree (subsp. europaea) and five other subspecies (laperrinei, guanchica, maroccana, cerasiformis, and cuspidata) distributed across the Old World, including Macaronesian islands. The evolutionary history of this monophyletic group (O. europaea complex) has revealed a reticulated scenario involving hybridization and polyploidization events, leading to the presence of a polyploid series associated with the subspecies. However, how the polyploids originated, and how the different subspecies contributed to the domestication of the cultivated olive are questions still debated. Tracing the recent evolution and genetic diversification of the species is key for the management and preservation of its genetic resources. To study the recent history of the O. europaea complex, we compared newly sequenced and available genomes for 27 individuals representing the six subspecies. RESULTS Our results show discordance between current subspecies distributions and phylogenomic patterns, which support intricate biogeographic patterns. The subspecies guanchica, restricted to the Canary Islands, is closely related to subsp. europaea, and shows a high genetic diversity. The subsp. laperrinei, restricted now to high mountains of the Sahara desert, and the Canarian subsp. guanchica contributed to the formation of the allotetraploid subsp. cerasiformis (Madeira islands) and the allohexaploid subsp. maroccana (western Sahara region). Our phylogenomic data support the recognition of one more taxon (subsp. ferruginea) for the Asian populations, which is clearly segregated from the African subsp. cuspidata. CONCLUSIONS In sum, the O. europaea complex underwent several processes of hybridization, polyploidy, and geographical isolation resulting in seven independent lineages with certain morphological traits recognised into subspecies.
Collapse
Affiliation(s)
- Irene Julca
- Barcelona Supercomputing Centre (BSC-CNS), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Pablo Vargas
- Department of Biodiversity and Conservation, Real Jardín Botánico de Madrid, Calle Claudio Moyano 1, 28014, Madrid, Spain
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), Plaça Eusebi Güell, 1-3, 08034, Barcelona, Spain.
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
47
|
Hurley LD, Lee H, Wade G, Simcox J, Engin F. Ormdl3 regulation of specific ceramides is dispensable for mouse β-cell function and glucose homeostasis under obesogenic conditions. Front Endocrinol (Lausanne) 2023; 14:1170461. [PMID: 37124760 PMCID: PMC10140491 DOI: 10.3389/fendo.2023.1170461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Chronic elevation of sphingolipids contributes to β-cell failure. ORMDL3 has been identified as a key regulator of sphingolipid homeostasis, however, its function in pancreatic β-cell pathophysiology remains unclear. Here, we generated a mouse model lacking Ormdl3 within pancreatic β-cells (Ormdl3 β-/-). We show that loss of β-cell Ormdl3 does not alter glucose tolerance, insulin sensitivity, insulin secretion, islet morphology, or cellular ceramide levels on standard chow diet. When challenged with a high fat diet, while Ormdl3 β-/- mice did not exhibit any alteration in metabolic parameters or islet architecture, lipidomics analysis revealed significantly higher levels of very long chain ceramides in their islets. Taken together, our results reveal that loss of Ormdl3 alone is not sufficient to impinge upon β-cell function or whole-body glucose and insulin homeostasis, however, β-cell-specific loss of Ormdl3 does significantly alter levels of specific sphingolipid species in islets upon high fat feeding.
Collapse
Affiliation(s)
- Liam D. Hurley
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Hugo Lee
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Gina Wade
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Judith Simcox
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Feyza Engin
- Department of Biomolecular Chemistry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
48
|
Ikushiro H, Murakami T, Takahashi A, Katayama A, Sawai T, Goto H, Koolath S, Murai Y, Monde K, Miyahara I, Kamiya N, Yano T. Structural insights into the substrate recognition of serine palmitoyltransferase from Sphingobacterium multivorum. J Biol Chem 2023; 299:104684. [PMID: 37030501 DOI: 10.1016/j.jbc.2023.104684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/28/2023] [Accepted: 04/01/2023] [Indexed: 04/10/2023] Open
Abstract
Serine palmitoyltransferase (SPT) is a key enzyme of sphingolipid biosynthesis, which catalyzes the pyridoxal-5'-phosphate-dependent decarboxylative condensation reaction of L-serine (L-Ser) and palmitoyl-CoA (PalCoA) to form 3-ketodihydrosphingosine called long chain base (LCB). SPT is also able to metabolize L-alanine (L-Ala) and glycine (Gly), albeit with much lower efficiency. Human SPT is a membrane-bound large protein complex containing SPTLC1/SPTLC2 heterodimer as the core subunits, and it is known that mutations of the SPTLC1/SPTLC2 genes increase the formation of deoxy-type of LCBs derived from L-Ala and Gly to cause some neurodegenerative diseases. In order to study the substrate recognition of SPT, we examined the reactivity of Sphingobacterium multivorum SPT on various amino acids in the presence of PalCoA. The S. multivorum SPT could convert not only L-Ala and Gly but also L-homoserine, in addition to L-Ser, into the corresponding LCBs. Furthermore, we obtained high-quality crystals of the ligand-free form and the binary complexes with a series of amino acids, including a nonproductive amino acid, L-threonine, and determined the structures at 1.40-1.55 Å resolutions. The S. multivorum SPT accommodated various amino acid substrates through subtle rearrangements of the active-site amino acid residues and water molecules. It was also suggested that non-active-site residues mutated in the human SPT genes might indirectly influence the substrate specificity by affecting the hydrogen-bonding networks involving the bound substrate, water molecules, and amino acid residues in the active site of this enzyme. Collectively, our results highlight SPT structural features affecting substrate specificity for this stage of sphingolipid biosynthesis.
Collapse
Affiliation(s)
- Hiroko Ikushiro
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| | - Taiki Murakami
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka, Osaka 558-8585, Japan
| | - Aya Takahashi
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka, Osaka 558-8585, Japan
| | - Asuka Katayama
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka, Osaka 558-8585, Japan
| | - Taiki Sawai
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Haruna Goto
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Sajeer Koolath
- Frontier Research Center for Advanced Material and Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita21 Nishi11, Sapporo, Hokkaido 001-0021, JAPAN
| | - Yuta Murai
- Frontier Research Center for Advanced Material and Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita21 Nishi11, Sapporo, Hokkaido 001-0021, JAPAN
| | - Kenji Monde
- Frontier Research Center for Advanced Material and Life Science, Faculty of Advanced Life Science, Hokkaido University, Kita21 Nishi11, Sapporo, Hokkaido 001-0021, JAPAN
| | - Ikuko Miyahara
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka, Osaka 558-8585, Japan
| | - Nobuo Kamiya
- Department of Chemistry, Graduate School of Science, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka, Osaka 558-8585, Japan; Research Center for Artificial Photosynthesis, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka, Osaka 558-8585, Japan
| | - Takato Yano
- Department of Biochemistry, Faculty of Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| |
Collapse
|
49
|
Liu P, Xie T, Wu X, Han G, Gupta SD, Zhang Z, Yue J, Dong F, Gable K, Niranjanakumari S, Li W, Wang L, Liu W, Yao R, Cahoon EB, Dunn TM, Gong X. Mechanism of sphingolipid homeostasis revealed by structural analysis of Arabidopsis SPT-ORM1 complex. SCIENCE ADVANCES 2023; 9:eadg0728. [PMID: 36989369 PMCID: PMC10058238 DOI: 10.1126/sciadv.adg0728] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/02/2023] [Indexed: 06/19/2023]
Abstract
The serine palmitoyltransferase (SPT) complex catalyzes the first and rate-limiting step in sphingolipid biosynthesis in all eukaryotes. ORM/ORMDL proteins are negative regulators of SPT that respond to cellular sphingolipid levels. However, the molecular basis underlying ORM/ORMDL-dependent homeostatic regulation of SPT is not well understood. We determined the cryo-electron microscopy structure of Arabidopsis SPT-ORM1 complex, composed of LCB1, LCB2a, SPTssa, and ORM1, in an inhibited state. A ceramide molecule is sandwiched between ORM1 and LCB2a in the cytosolic membrane leaflet. Ceramide binding is critical for the ORM1-dependent SPT repression, and dihydroceramides and phytoceramides differentially affect this repression. A hybrid β sheet, formed by the amino termini of ORM1 and LCB2a and induced by ceramide binding, stabilizes the amino terminus of ORM1 in an inhibitory conformation. Our findings provide mechanistic insights into sphingolipid homeostatic regulation via the binding of ceramide to the SPT-ORM/ORMDL complex that may have implications for plant-specific processes such as the hypersensitive response for microbial pathogen resistance.
Collapse
Affiliation(s)
- Peng Liu
- Department of Chemical Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Tian Xie
- Department of Chemical Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xinyue Wu
- Department of Chemical Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Gongshe Han
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sita D. Gupta
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Zike Zhang
- Department of Chemical Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jian Yue
- Department of Chemical Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Feitong Dong
- Department of Chemical Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Kenneth Gable
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Somashekarappa Niranjanakumari
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Wanyuan Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Plant Functional Genomics and Developmental Regulation, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Lin Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Plant Functional Genomics and Developmental Regulation, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Wenchen Liu
- Department of Chemical Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Ruifeng Yao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Plant Functional Genomics and Developmental Regulation, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Edgar B. Cahoon
- Center for Plant Science Innovation and Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Teresa M. Dunn
- Department of Biochemistry and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Xin Gong
- Department of Chemical Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
50
|
van Echten-Deckert G. The role of sphingosine 1-phosphate metabolism in brain health and disease. Pharmacol Ther 2023; 244:108381. [PMID: 36907249 DOI: 10.1016/j.pharmthera.2023.108381] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023]
Abstract
Lipids are essential structural and functional components of the central nervous system (CNS). Sphingolipids are ubiquitous membrane components which were discovered in the brain in the late 19th century. In mammals, the brain contains the highest concentration of sphingolipids in the body. Sphingosine 1-phosphate (S1P) derived from membrane sphingolipids evokes multiple cellular responses which, depending on its concentration and localization, make S1P a double-edged sword in the brain. In the present review we highlight the role of S1P in brain development and focus on the often contrasting findings regarding its contributions to the initiation, progression and potential recovery of different brain pathologies, including neurodegeneration, multiple sclerosis (MS), brain cancers, and psychiatric illnesses. A detailed understanding of the critical implications of S1P in brain health and disease may open the door for new therapeutic options. Thus, targeting S1P-metabolizing enzymes and/or signaling pathways might help overcome, or at least ameliorate, several brain illnesses.
Collapse
|