1
|
Zaidalkilani AT, Al-Kuraishy HM, Fahad EH, Al-Gareeb AI, Elewa YHA, Zahran MH, Alexiou A, Papadakis M, Al-Farga A, Batiha GES. Autophagy modulators in type 2 diabetes: A new perspective. J Diabetes 2024; 16:e70010. [PMID: 39676616 DOI: 10.1111/1753-0407.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 05/31/2024] [Accepted: 08/27/2024] [Indexed: 12/17/2024] Open
Abstract
Type 2 diabetes (T2D) is a chronic metabolic disorder caused by defective insulin signaling, insulin resistance, and impairment of insulin secretion. Autophagy is a conserved lysosomal-dependent catabolic cellular pathway involved in the pathogenesis of T2D and its complications. Basal autophagy regulates pancreatic β-cell function by enhancing insulin release and peripheral insulin sensitivity. Therefore, defective autophagy is associated with impairment of pancreatic β-cell function and the development of insulin rersistance (IR). However, over-activated autophagy increases apoptosis of pancreatic β-cells leading to pancreatic β-cell dysfunction. Hence, autophagy plays a double-edged sword role in T2D. Therefore, the use of autophagy modulators including inhibitors and activators may affect the pathogenesis of T2D. Hence, this review aims to clarify the potential role of autophagy inhibitors and activators in T2D.
Collapse
Affiliation(s)
- Ayah Talal Zaidalkilani
- Department of Nutrition, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq
| | - Esraa H Fahad
- Department of Pharmacology and Toxicology, College of Pharmacy, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
- Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mahmoud Hosny Zahran
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, Greece
- Department of Research & Development, AFNP Med, Wien, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Wuppertal, Germany
| | - Ammar Al-Farga
- Department of Biochemistry, College of Science University of Jeddah, Jeddah, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhur University, Damanhur, AlBeheira, Egypt
| |
Collapse
|
2
|
Kuo A, Hla T. Regulation of cellular and systemic sphingolipid homeostasis. Nat Rev Mol Cell Biol 2024; 25:802-821. [PMID: 38890457 DOI: 10.1038/s41580-024-00742-y] [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] [Accepted: 04/30/2024] [Indexed: 06/20/2024]
Abstract
One hundred and fifty years ago, Johann Thudichum described sphingolipids as unusual "Sphinx-like" lipids from the brain. Today, we know that thousands of sphingolipid molecules mediate many essential functions in embryonic development and normal physiology. In addition, sphingolipid metabolism and signalling pathways are dysregulated in a wide range of pathologies, and therapeutic agents that target sphingolipids are now used to treat several human diseases. However, our understanding of sphingolipid regulation at cellular and organismal levels and their functions in developmental, physiological and pathological settings is rudimentary. In this Review, we discuss recent advances in sphingolipid pathways in different organelles, how secreted sphingolipid mediators modulate physiology and disease, progress in sphingolipid-targeted therapeutic and diagnostic research, and the trans-cellular sphingolipid metabolic networks between microbiota and mammals. Advances in sphingolipid biology have led to a deeper understanding of mammalian physiology and may lead to progress in the management of many diseases.
Collapse
Affiliation(s)
- Andrew Kuo
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Acun AD, Kantar D. Modulation of oxidative stress and apoptosis by alteration of bioactive lipids in the pancreas, and effect of zinc chelation in a rat model of Alzheimer's disease. J Trace Elem Med Biol 2024; 85:127480. [PMID: 38875759 DOI: 10.1016/j.jtemb.2024.127480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
INTRODUCTION Increasing epidemiological evidence highlights the association between systemic insulin resistance and Alzheimer's disease (AD). It is known that peripheral insulin resistance in the early stages of AD precedes and is a precursor to amyloid-β (Aβ) deposition. Although it is known that improving the CNS insulin sensitivity of AD patients is an important therapeutic goal and that the majority of insulin in the brain comes from the periphery, there has been little attention to the changes that occur in the pancreatic tissue of AD patients. Therefore, it is crucial to elucidate the mechanisms affecting insulin resistance in pancreatic tissue in AD. It is known that zinc (Zn2+) chelation is effective in reducing peripheral insulin resistance, cell apoptosis, cell death, and oxidative stress. OBJECTIVE It was aimed to determine the changes in bioactive lipids, amylin (AIPP), oxidative stress and apoptosis in pancreatic cells in the early stages of Alzheimer's disease. The main aim is to reveal the therapeutic effect of the Cyclo-Z agent on these changes seen in the pancreas due to AD disease. METHODS AD and ADC rats were intracerebroventricular (i.c.v.) Aβ1-42 oligomers. Cyclo-Z gavage was applied to ADC and SHC rats for 21 days. First of all, the effects of AIPP, bioactive ceramides, apoptosis and oxidative stress on the pancreatic tissue of AD group rats were evaluated. Then, the effect of Cyclo-Z treatment on these was examined. ELISA kit was used in biochemical analyses. RESULTS AIPP and ceramide (CER) levels and CER/ sphingosine-1 phosphate (S1P) ratio were increased in the pancreatic tissue of AD rats. It also increased the level of CER kinase (CERK), which is known to increase the concentration of CER 1-phosphate (C1P), which is known to be toxic to cells in the presence of excessive CER concentration. Due to the increase in CER level, it was observed that apoptosis and oxidative stress increased in the pancreatic cells of AD group rats. CONCLUSION Cyclo-Z, which has Zn2+ chelating properties, reduced AD model rats' AIPP level and oxidative stress and could prevent pancreatic apoptosis. Similar therapeutic effects were not observed in the pancreatic tissue of Cyclo-Z administered to the SH group. For this reason, it is thought that Cyclo-Z agent may have a therapeutic effect on the peripheral hyperinsulinemia observed in the early stages of AD disease and the resulting low amount of insulin transported to the brain, by protecting pancreatic cells from apoptosis and oxidative stress by regulating their bioactive metabolites.
Collapse
Affiliation(s)
- Alev Duygu Acun
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Arapsuyu, Antalya 07070, Turkey.
| | - Deniz Kantar
- Department of Biophysics, Faculty of Medicine, Akdeniz University, Arapsuyu, Antalya 07070, Turkey
| |
Collapse
|
4
|
Jia W, Yuan J, Zhang J, Li S, Lin W, Cheng B. Bioactive sphingolipids as emerging targets for signal transduction in cancer development. Biochim Biophys Acta Rev Cancer 2024; 1879:189176. [PMID: 39233263 DOI: 10.1016/j.bbcan.2024.189176] [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: 06/06/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Sphingolipids, crucial components of cellular membranes, play a vital role in maintaining cellular structure and signaling integrity. Disruptions in sphingolipid metabolism are increasingly implicated in cancer development. Key bioactive sphingolipids, such as ceramides, sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), and glycosphingolipids, profoundly impact tumor biology. They influence the behavior of tumor cells, stromal cells, and immune cells, affecting tumor aggressiveness, angiogenesis, immune modulation, and extracellular matrix remodeling. Furthermore, abnormal expression of sphingolipids and their metabolizing enzymes modulates the secretion of tumor-derived extracellular vesicles (TDEs), which are key players in creating an immunosuppressive tumor microenvironment, remodeling the extracellular matrix, and facilitating oncogenic signaling within in situ tumors and distant pre-metastatic niches (PMNs). Understanding the role of sphingolipids in the biogenesis of tumor-derived extracellular vesicles (TDEs) and their bioactive contents can pave the way for new biomarkers in cancer diagnosis and prognosis, ultimately enhancing comprehensive tumor treatment strategies.
Collapse
Affiliation(s)
- Wentao Jia
- Department of General Practice, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China
| | - Jiaying Yuan
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jinbo Zhang
- Department of Pharmacy, Tianjin Rehabilitation and Recuperation Center, Joint Logistics Support Force, Tianjin 300000, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Wanfu Lin
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| |
Collapse
|
5
|
Yadav P, Rana K, Chakraborty R, Khan A, Mehta D, Jain D, Aggarwal B, Jha SK, Dasgupta U, Bajaj A. Engineered nanomicelles targeting proliferation and angiogenesis inhibit tumour progression by impairing the synthesis of ceramide-1-phosphate. NANOSCALE 2024; 16:10350-10365. [PMID: 38739006 DOI: 10.1039/d3nr04806c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Tumour cells secrete various proangiogenic factors like VEGF, PDGF, and EGF that result in the formation of highly vascularized tumours with an immunosuppressive tumour microenvironment. As tumour growth and metastasis are highly dependent on angiogenesis, targeting tumour vasculature along with rapidly dividing tumour cells is a potential approach for cancer treatment. Here, we specifically engineered sub-100 sized nanomicelles (DTX-CA4 NMs) targeting proliferation and angiogenesis using an esterase-sensitive phosphocholine-tethered docetaxel conjugate of lithocholic acid (LCA) (PC-LCA-DTX) and a poly(ethylene glycol) (PEG) derivative of an LCA-combretastatin A4 conjugate (PEG-LCA-CA4). DTX-CA4 NMs effectively inhibit the tumour growth in syngeneic (CT26) and xenograft (HCT116) colorectal cancer models, inhibit tumour recurrence, and enhance the percentage survival in comparison with individual drug-loaded NMs. DTX-CA4 NMs enhance the T cell-mediated anti-tumour immune response and DTX-CA4 NMs in combination with an immune checkpoint inhibitor, anti-PDL1 antibody, enhance the anti-tumour response. We additionally showed that DTX-CA4 NMs effectively attenuate the production of ceramide-1-phosphate, a key metabolite of the sphingolipid pathway, by downregulating the expression of ceramide kinase at both transcriptional and translational levels. Therefore, this study presents the engineering of effective DTX-CA4 NMs for targeting the tumour microenvironment that can be explored further for clinical applications.
Collapse
Affiliation(s)
- Poonam Yadav
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India.
| | - Kajal Rana
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India.
| | - Ruchira Chakraborty
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India.
| | - Ali Khan
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon-122413, Haryana, India
| | - Devashish Mehta
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon-122413, Haryana, India
| | - Dolly Jain
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India.
| | - Bharti Aggarwal
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India.
| | - Somesh K Jha
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India.
| | - Ujjaini Dasgupta
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Manesar, Gurgaon-122413, Haryana, India
| | - Avinash Bajaj
- Laboratory of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone Faridabad-Gurgaon Expressway, Faridabad-121001, Haryana, India.
| |
Collapse
|
6
|
Tolerico M, Merscher S, Fornoni A. Normal and Dysregulated Sphingolipid Metabolism: Contributions to Podocyte Injury and Beyond. Cells 2024; 13:890. [PMID: 38891023 PMCID: PMC11171506 DOI: 10.3390/cells13110890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/09/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024] Open
Abstract
Podocyte health is vital for maintaining proper glomerular filtration in the kidney. Interdigitating foot processes from podocytes form slit diaphragms which regulate the filtration of molecules through size and charge selectivity. The abundance of lipid rafts, which are ordered membrane domains rich in cholesterol and sphingolipids, near the slit diaphragm highlights the importance of lipid metabolism in podocyte health. Emerging research shows the importance of sphingolipid metabolism to podocyte health through structural and signaling roles. Dysregulation in sphingolipid metabolism has been shown to cause podocyte injury and drive glomerular disease progression. In this review, we discuss the structure and metabolism of sphingolipids, as well as their role in proper podocyte function and how alterations in sphingolipid metabolism contributes to podocyte injury and drives glomerular disease progression.
Collapse
Affiliation(s)
| | - Sandra Merscher
- Peggy and Harold Katz Family Drug Discovery Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Alessia Fornoni
- Peggy and Harold Katz Family Drug Discovery Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| |
Collapse
|
7
|
Sosa-Acosta P, Quiñones-Vega M, Guedes JDS, Rocha D, Guida L, Vasconcelos Z, Nogueira FCS, Domont GB. Multiomics Approach Reveals Serum Biomarker Candidates for Congenital Zika Syndrome. J Proteome Res 2024; 23:1200-1220. [PMID: 38390744 DOI: 10.1021/acs.jproteome.3c00677] [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: 02/24/2024]
Abstract
The Zika virus (ZIKV) can be vertically transmitted, causing congenital Zika syndrome (CZS) in fetuses. ZIKV infection in early gestational trimesters increases the chances of developing CZS. This syndrome involves several pathologies with a complex diagnosis. In this work, we aim to identify biological processes and molecular pathways related to CZS and propose a series of putative protein and metabolite biomarkers for CZS prognosis in early pregnancy trimesters. We analyzed serum samples of healthy pregnant women and ZIKV-infected pregnant women bearing nonmicrocephalic and microcephalic fetuses. A total of 1090 proteins and 512 metabolites were identified by bottom-up proteomics and untargeted metabolomics, respectively. Univariate and multivariate statistical approaches were applied to find CZS differentially abundant proteins (DAP) and metabolites (DAM). Enrichment analysis (i.e., biological processes and molecular pathways) of the DAP and the DAM allowed us to identify the ECM organization and proteoglycans, amino acid metabolism, and arachidonic acid metabolism as CZS signatures. Five proteins and four metabolites were selected as CZS biomarker candidates. Serum multiomics analysis led us to propose nine putative biomarkers for CZS prognosis with high sensitivity and specificity.
Collapse
Affiliation(s)
- Patricia Sosa-Acosta
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
- Laboratory of Proteomics (LabProt), LADETEC, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
- Precision Medicine Research Center, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Mauricio Quiñones-Vega
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
- Laboratory of Proteomics (LabProt), LADETEC, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
- Precision Medicine Research Center, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Jéssica de S Guedes
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
- Laboratory of Proteomics (LabProt), LADETEC, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
- Precision Medicine Research Center, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Danielle Rocha
- Fernandes Figueira Institute, Fiocruz, Rio de Janeiro 22250-020, Brazil
| | - Letícia Guida
- Fernandes Figueira Institute, Fiocruz, Rio de Janeiro 22250-020, Brazil
| | | | - Fábio C S Nogueira
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
- Laboratory of Proteomics (LabProt), LADETEC, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
- Precision Medicine Research Center, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Gilberto B Domont
- Proteomics Unit, Department of Biochemistry, Institute of Chemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
- Precision Medicine Research Center, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| |
Collapse
|
8
|
Xiao H, Yin D, Du L, Li G, Lin J, Fang C, Shen S, Xiao G, Fang R. Effects of pork sausage on intestinal microecology and metabolism in mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:3413-3427. [PMID: 38111159 DOI: 10.1002/jsfa.13227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/01/2023] [Accepted: 12/16/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Processed meat, as an important part of the human diet, has been recognized as a carcinogen by the International Agency for Research on Cancer (IARC). Although numerous epidemiological reports supported the IARC's view, the relevant evidence of a direct association between processed meat and carcinogenicity has been insufficient and the mechanism has been unclear. This study aims to investigate the effects of pork sausage (as a representative example of processed meat) intake on gut microbial communities and metabolites of mice. Microbial communities and metabolites from all groups were analyzed using 16S rRNA gene sequencing and Ultra performance liquid chromatography-quadrupole-time of flight-mass spectrometer (UPLC-Q-TOF/MS), respectively. RESULTS The levels of Bacteroidetes, Bacteroides, Alloprevotella, Lactobacillus, Prevotella_9, Lachnospiraceae_NK4A136_group, Alistipes, Blautia, Proteobacteria, Firmicutes, Allobaculum, Helicobacter, Desulfovibrio, Clostridium_sensu_stricto_1, Ruminococcaceae_UCG-014, Lachnospiraceae_UCG-006 and Streptococcus (P < 0.05) were obviously altered in the mice fed a pork sausage diet. Twenty-seven metabolites from intestinal content samples and fourteen matabolites from whole blood samples were identified as potential biomarkers from multivariate analysis, including Phosphatidic acid (PA), Sphingomyelin (SM), Lysophosphatidylcholine (LysoPC), Diglyceride (DG), D-maltose, N-acylamides and so forth. The significant changes in these biomarkers demonstrate metabonomic variations in pork sausage treated rats, especially carbohydrate metabolism, lipid metabolism, and amino acid metabolism. CONCLUSION The present study provided evidence that a processed meat diet can increase the risk of colorectal cancer and other diseases significantly by altering the microbial community structure and disrupting the body's metabolic pathways. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Hailong Xiao
- Key Laboratory of Agricultural Products Chemical and Biological Processing Technology, Zhejiang University of Science and Technology, Hangzhou, China
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Danhan Yin
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Lidan Du
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Gaotian Li
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Jie Lin
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Chenyu Fang
- Hangzhou Institute for Food and Drug Control, Hangzhou, China
| | - Shaolin Shen
- Hangzhou Xiaoshan Institute of Measurement for Quality and Technique Supervision, Hangzhou, China
| | - Gongnian Xiao
- Key Laboratory of Agricultural Products Chemical and Biological Processing Technology, Zhejiang University of Science and Technology, Hangzhou, China
| | - Ruosi Fang
- Key Laboratory of Agricultural Products Chemical and Biological Processing Technology, Zhejiang University of Science and Technology, Hangzhou, China
| |
Collapse
|
9
|
Choudhary V, Choudhary M, Bollag WB. Exploring Skin Wound Healing Models and the Impact of Natural Lipids on the Healing Process. Int J Mol Sci 2024; 25:3790. [PMID: 38612601 PMCID: PMC11011291 DOI: 10.3390/ijms25073790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/14/2024] Open
Abstract
Cutaneous wound healing is a complex biological process involving a series of well-coordinated events aimed at restoring skin integrity and function. Various experimental models have been developed to study the mechanisms underlying skin wound repair and to evaluate potential therapeutic interventions. This review explores the diverse array of skin wound healing models utilized in research, ranging from rodent excisional wounds to advanced tissue engineering constructs and microfluidic platforms. More importantly, the influence of lipids on the wound healing process is examined, emphasizing their role in enhancing barrier function restoration, modulating inflammation, promoting cell proliferation, and promoting remodeling. Lipids, such as phospholipids, sphingolipids, and ceramides, play crucial roles in membrane structure, cell signaling, and tissue repair. Understanding the interplay between lipids and the wound microenvironment provides valuable insights into the development of novel therapeutic strategies for promoting efficient wound healing and tissue regeneration. This review highlights the significance of investigating skin wound healing models and elucidating the intricate involvement of lipids in the healing process, offering potential avenues for improving clinical outcomes in wound management.
Collapse
Affiliation(s)
- Vivek Choudhary
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (V.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Mrunal Choudhary
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (V.C.)
| | - Wendy B. Bollag
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (V.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Department of Dermatology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
10
|
Engin AB. Mechanism of Obesity-Related Lipotoxicity and Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:131-166. [PMID: 39287851 DOI: 10.1007/978-3-031-63657-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The link between cellular exposure to fatty acid species and toxicity phenotypes remains poorly understood. However, structural characterization and functional profiling of human plasma free fatty acids (FFAs) analysis has revealed that FFAs are located either in the toxic cluster or in the cluster that is transcriptionally responsive to lipotoxic stress and creates genetic risk factors. Genome-wide short hairpin RNA screen has identified more than 350 genes modulating lipotoxicity. Hypertrophic adipocytes in obese adipose are both unable to expand further to store excess lipids in the diet and are resistant to the antilipolytic action of insulin. In addition to lipolysis, the inability of packaging the excess lipids into lipid droplets causes circulating fatty acids to reach toxic levels in non-adipose tissues. Deleterious effects of accumulated lipid in non-adipose tissues are known as lipotoxicity. Although triglycerides serve a storage function for long-chain non-esterified fatty acid and their products such as ceramide and diacylglycerols (DAGs), overloading of palmitic acid fraction of saturated fatty acids (SFAs) raises ceramide levels. The excess DAG and ceramide load create harmful effects on multiple organs and systems, inducing chronic inflammation in obesity. Thus, lipotoxic inflammation results in β cells death and pancreatic islets dysfunction. Endoplasmic reticulum stress stimuli induce lipolysis by activating cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) and extracellular signal-regulated kinase (Erk) 1/2 signaling in adipocytes. However, palmitic acid-induced endoplasmic reticulum stress-c-Jun N-terminal kinase (JNK)-autophagy axis in hypertrophic adipocytes is a pro-survival mechanism against endoplasmic reticulum stress and cell death induced by SFAs. Endoplasmic reticulum-localized acyl-coenzyme A (CoA): glycerol-3-phosphate acyltransferase (GPAT) enzymes are mediators of lipotoxicity, and inhibiting these enzymes has therapeutic potential for lipotoxicity. Lipotoxicity increases the number of autophagosomes, which engulf palmitic acid, and thus suppress the autophagic turnover. Fatty acid desaturation promotes palmitate detoxification and storages into triglycerides. As therapeutic targets of glucolipotoxicity, in addition to caloric restriction and exercise, there are four different pharmacological approaches, which consist of metformin, glucagon-like peptide 1 (GLP-1) receptor agonists, peroxisome proliferator-activated receptor-gamma (PPARγ) ligands thiazolidinediones, and chaperones are still used in clinical practice. Furthermore, induction of the brown fat-like phenotype with the mixture of eicosapentanoic acid and docosahexaenoic acid appears as a potential therapeutic application for treatment of lipotoxicity.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| |
Collapse
|
11
|
Peng X, Du J, Wang Y. Metabolic signatures in post-myocardial infarction heart failure, including insights into prediction, intervention, and prognosis. Biomed Pharmacother 2024; 170:116079. [PMID: 38150879 DOI: 10.1016/j.biopha.2023.116079] [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: 09/19/2023] [Revised: 12/09/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023] Open
Abstract
Heart failure (HF) is a prevalent long-term complication of myocardial infarction (MI). The incidence of post-MI HF is high, and patients with the condition have a poor prognosis. Accurate identification of individuals at high risk for post-MI HF is crucial for implementation of a protective and ideally personalized strategy to prevent fatal events. Post-MI HF is characterized by adverse cardiac remodeling, which results from metabolic changes in response to long-term ischemia. Moreover, various risk factors, including genetics, diet, and obesity, can influence metabolic pathways in patients. This review focuses on the metabolic signatures of post-MI HF that could serve as non-invasive biomarkers for early identification in high-risk populations. We also explore how metabolism participates in the pathophysiology of post-MI HF. Furthermore, we discuss the potential of metabolites as novel targets for treatment of post-MI HF and as biomarkers for prognostic evaluation. It is expected to provide valuable suggestions for the clinical prevention and treatment of post-MI HF from a metabolic perspective.
Collapse
Affiliation(s)
- Xueyan Peng
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China; Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China
| | - Jie Du
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China; Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China.
| | - Yuan Wang
- Beijing Collaborative Innovation Centre for Cardiovascular Disorders, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China; Key Laboratory of Remodeling-related Cardiovascular Diseases, Ministry of Education, Beijing 100029, China; Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; Beijing Institute of Heart, Lung and Blood Vessel Disease, No. 2 Anzhen Road, Chaoyang District, Beijing 100029, China.
| |
Collapse
|
12
|
Pawar A, Zabetakis I, Gavankar T, Lordan R. Milk polar lipids: Untapped potential for pharmaceuticals and nutraceuticals. PHARMANUTRITION 2023. [DOI: 10.1016/j.phanu.2023.100335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
|
13
|
Rogers JR, Geissler PL. Ceramide-1-phosphate transfer protein enhances lipid transport by disrupting hydrophobic lipid-membrane contacts. PLoS Comput Biol 2023; 19:e1010992. [PMID: 37036851 PMCID: PMC10085062 DOI: 10.1371/journal.pcbi.1010992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 03/03/2023] [Indexed: 04/11/2023] Open
Abstract
Cellular distributions of the sphingolipid ceramide-1-phosphate (C1P) impact essential biological processes. C1P levels are spatiotemporally regulated by ceramide-1-phosphate transfer protein (CPTP), which efficiently shuttles C1P between organelle membranes. Yet, how CPTP rapidly extracts and inserts C1P into a membrane remains unknown. Here, we devise a multiscale simulation approach to elucidate biophysical details of CPTP-mediated C1P transport. We find that CPTP binds a membrane poised to extract and insert C1P and that membrane binding promotes conformational changes in CPTP that facilitate C1P uptake and release. By significantly disrupting a lipid's local hydrophobic environment in the membrane, CPTP lowers the activation free energy barrier for passive C1P desorption and enhances C1P extraction from the membrane. Upon uptake of C1P, further conformational changes may aid membrane unbinding in a manner reminiscent of the electrostatic switching mechanism used by other lipid transfer proteins. Insertion of C1P into an acceptor membrane, eased by a decrease in membrane order by CPTP, restarts the transfer cycle. Most notably, we provide molecular evidence for CPTP's ability to catalyze C1P extraction by breaking hydrophobic C1P-membrane contacts with compensatory hydrophobic lipid-protein contacts. Our work, thus, provides biophysical insights into how CPTP efficiently traffics C1P between membranes to maintain sphingolipid homeostasis and, additionally, presents a simulation method aptly suited for uncovering the catalytic mechanisms of other lipid transfer proteins.
Collapse
Affiliation(s)
- Julia R Rogers
- Department of Chemistry, University of California, Berkeley, California, United States of America
| | - Phillip L Geissler
- Department of Chemistry, University of California, Berkeley, California, United States of America
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| |
Collapse
|
14
|
Neuroinflammation, Energy and Sphingolipid Metabolism Biomarkers Are Revealed by Metabolic Modeling of Autistic Brains. Biomedicines 2023; 11:biomedicines11020583. [PMID: 36831124 PMCID: PMC9953696 DOI: 10.3390/biomedicines11020583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/18/2023] Open
Abstract
Autism spectrum disorders (ASD) are a heterogeneous group of neurodevelopmental disorders generally characterized by repetitive behaviors and difficulties in communication and social behavior. Despite its heterogeneous nature, several metabolic dysregulations are prevalent in individuals with ASD. This work aims to understand ASD brain metabolism by constructing an ASD-specific prefrontal cortex genome-scale metabolic model (GEM) using transcriptomics data to decipher novel neuroinflammatory biomarkers. The healthy and ASD-specific models are compared via uniform sampling to identify ASD-exclusive metabolic features. Noticeably, the results of our simulations and those found in the literature are comparable, supporting the accuracy of our reconstructed ASD model. We identified that several oxidative stress, mitochondrial dysfunction, and inflammatory markers are elevated in ASD. While oxidative phosphorylation fluxes were similar for healthy and ASD-specific models, and the fluxes through the pathway were nearly undisturbed, the tricarboxylic acid (TCA) fluxes indicated disruptions in the pathway. Similarly, the secretions of mitochondrial dysfunction markers such as pyruvate are found to be higher, as well as the activities of oxidative stress marker enzymes like alanine and aspartate aminotransferases (ALT and AST) and glutathione-disulfide reductase (GSR). We also detected abnormalities in the sphingolipid metabolism, which has been implicated in many inflammatory and immune processes, but its relationship with ASD has not been thoroughly explored in the existing literature. We suggest that important sphingolipid metabolites, such as sphingosine-1-phosphate (S1P), ceramide, and glucosylceramide, may be promising biomarkers for the diagnosis of ASD and provide an opportunity for the adoption of early intervention for young children.
Collapse
|
15
|
Horse-Derived Ceramide Accentuates Glucosylceramide Synthase and Ceramide Synthase 3 by Activating PPARβ/δ and/or PPARγ to Stimulate Ceramide Synthesis. Biomedicines 2023; 11:biomedicines11020548. [PMID: 36831084 PMCID: PMC9953238 DOI: 10.3390/biomedicines11020548] [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: 12/05/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Horse-derived ceramide (HC), which contains galactosylceramides as its main component, significantly improves skin symptoms when applied topically to patients with atopic dermatitis. We speculated that efficacy resulted from the amelioration of epidermal ceramide metabolism, and we characterized those effects using reconstructed human epidermal equivalents. Lipid analysis, RT-PCR and Western blotting revealed that HC significantly increased the total ceramide content of the stratum corneum (SC), accompanied by significantly increased gene and/or protein expression levels of ceramide synthase (CERS) 3, fatty acid elongase (ELOVL) 4, glucosylceramide synthase (GCS), β-glucocerebrosidase, sphingomyelin synthase and acid sphingomyelinase. Mechanistic analyses using cultures of primary human keratinocytes revealed the marked stimulatory effects of HC on the mRNA expression levels of CERS3, ELOVL4 and GCS under high calcium-derived differentiation conditions. Signaling analyses demonstrated that an antagonist of PPARβ/δ significantly abrogated the HC-stimulated mRNA expression levels of GCS, CERS3 and ELOVL4. GW9662, an antagonist of PPARγ, significantly abolished the HC-up-regulated mRNA expression levels of GCS and ELOVL4, but not of CERS3. These findings suggest that HC has the distinct potential to accentuate the expression of GCS, CERS3 and ELOVL4 via the activation of PPARβ/δ and/or PPARγ to accelerate ceramide synthesis in the SC.
Collapse
|
16
|
Huang C, Su L, Chen Y, Wu S, Sun R, Xu Q, Qiu X, Yang C, Kong X, Qin H, Zhao X, Jiang X, Wang K, Zhu Y, Wong PP. Ceramide kinase confers tamoxifen resistance in estrogen receptor-positive breast cancer by altering sphingolipid metabolism. Pharmacol Res 2023; 187:106558. [PMID: 36410675 DOI: 10.1016/j.phrs.2022.106558] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022]
Abstract
Dysregulated sphingolipid metabolism contributes to ER+ breast cancer progression and therapeutic response, whereas its underlying mechanism and contribution to tamoxifen resistance (TAMR) is unknown. Here, we establish sphingolipid metabolic enzyme CERK as a regulator of TAMR in breast cancer. Multi-omics analysis reveals an elevated CERK driven sphingolipid metabolic reprogramming in TAMR cells, while high CERK expression associates with worse patient prognosis in ER+ breast cancer. CERK overexpression confers tamoxifen resistance and promotes tumorigenicity in ER+ breast cancer cells. Knocking out CERK inhibits the orthotopic breast tumor growth of TAMR cells while rescuing their tamoxifen sensitivity. Mechanistically, the elevated EHF expression transcriptionally up-regulates CERK expression to prohibit tamoxifen-induced sphingolipid ceramide accumulation, which then inhibits tamoxifen-mediated repression on PI3K/AKT dependent cell proliferation and its driven p53/caspase-3 mediated apoptosis in TAMR cells. This work provides insight into the regulation of sphingolipid metabolism in tamoxifen resistance and identifies a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Cheng Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Liangping Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yitian Chen
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Sangqing Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Otolaryngology, Head and Neck Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ruipu Sun
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Qiuping Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaoyi Qiu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Ciqiu Yang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xiangzhan Kong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Hongquan Qin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xinbao Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xue Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| | - Yinghua Zhu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Laboratory Department, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China.
| | - Ping-Pui Wong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
17
|
Guzman G, Creek C, Farley S, Tafesse FG. Genetic Tools for Studying the Roles of Sphingolipids in Viral Infections. Methods Mol Biol 2022; 2610:1-16. [PMID: 36534277 DOI: 10.1007/978-1-0716-2895-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Sphingolipids are a critical family of membrane lipids with diverse functions in eukaryotic cells, and a growing body of literature supports that these lipids play essential roles during the lifecycles of viruses. While small molecule inhibitors of sphingolipid synthesis and metabolism are widely used, the advent of CRISPR-based genomic editing techniques allows for nuanced exploration into the manners in which sphingolipids influence various stages of viral infections. Here we describe some of these critical considerations needed in designing studies utilizing genomic editing techniques for manipulating the sphingolipid metabolic pathway, as well as the current body of literature regarding how viruses depend on the products of this pathway. Here, we highlight the ways in which sphingolipids affect viruses as these pathogens interact with and influence their host cell and describe some of the many open questions remaining in the field.
Collapse
Affiliation(s)
- Gaelen Guzman
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Cameron Creek
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Scotland Farley
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Fikadu G Tafesse
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
18
|
Presence of Ceramidase Activity in Electronegative LDL. Int J Mol Sci 2022; 24:ijms24010165. [PMID: 36613609 PMCID: PMC9820682 DOI: 10.3390/ijms24010165] [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: 09/05/2022] [Revised: 11/25/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Electronegative low-density lipoprotein (LDL(-)) is a minor modified fraction of human plasma LDL with several atherogenic properties. Among them is increased bioactive lipid mediator content, such as lysophosphatidylcholine (LPC), non-esterified fatty acids (NEFA), ceramide (Cer), and sphingosine (Sph), which are related to the presence of some phospholipolytic activities, including platelet-activating factor acetylhydrolase (PAF-AH), phospholipase C (PLC), and sphingomyelinase (SMase), in LDL(-). However, these enzymes' activities do not explain the increased Sph content, which typically derives from Cer degradation. In the present study, we analyzed the putative presence of ceramidase (CDase) activity, which could explain the increased Sph content. Thin layer chromatography (TLC) and lipidomic analysis showed that Cer, Sph, and NEFA spontaneously increased in LDL(-) incubated alone at 37 °C, in contrast with native LDL(+). An inhibitor of neutral CDase prevented the formation of Sph and, in turn, increased Cer content in LDL(-). In addition, LDL(-) efficiently degraded fluorescently labeled Cer (NBD-Cer) to form Sph and NEFA. These observations defend the existence of the CDase-like activity's association with LDL(-). However, neither the proteomic analysis nor the Western blot detected the presence of an enzyme with known CDase activity. Further studies are thus warranted to define the origin of the CDase-like activity detected in LDL(-).
Collapse
|
19
|
Oizumi H, Sugimura Y, Totsune T, Kawasaki I, Ohshiro S, Baba T, Kimpara T, Sakuma H, Hasegawa T, Kawahata I, Fukunaga K, Takeda A. Plasma sphingolipid abnormalities in neurodegenerative diseases. PLoS One 2022; 17:e0279315. [PMID: 36525454 PMCID: PMC9757566 DOI: 10.1371/journal.pone.0279315] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In recent years, there has been increasing evidence that several lipid metabolism abnormalities play an important role in the pathogenesis of neurodegenerative diseases. However, it is still unclear which lipid metabolism abnormalities play the most important role in neurodegenerative diseases. Plasma lipid metabolomics (lipidomics) has been shown to be an unbiased method that can be used to explore lipid metabolism abnormalities in neurodegenerative diseases. Plasma lipidomics in neurodegenerative diseases has been performed only in idiopathic Parkinson's disease (IPD) and Alzheimer's disease (AD), and comprehensive studies are needed to clarify the pathogenesis. METHODS In this study, we investigated plasma lipids using lipidomics in individuals with neurodegenerative diseases and healthy controls (CNs). Plasma lipidomics was evaluated by liquid chromatography-tandem mass spectrometry (LC-MS/MS) in those with IPD, dementia with Lewy bodies (DLB), multiple system atrophy (MSA), AD, and progressive supranuclear palsy (PSP) and CNs. RESULTS The results showed that (1) plasma sphingosine-1-phosphate (S1P) was significantly lower in all neurodegenerative disease groups (IPD, DLB, MSA, AD, and PSP) than in the CN group. (2) Plasma monohexylceramide (MonCer) and lactosylceramide (LacCer) were significantly higher in all neurodegenerative disease groups (IPD, DLB, MSA, AD, and PSP) than in the CN group. (3) Plasma MonCer levels were significantly positively correlated with plasma LacCer levels in all enrolled groups. CONCLUSION S1P, Glucosylceramide (GlcCer), the main component of MonCer, and LacCer are sphingolipids that are biosynthesized from ceramide. Recent studies have suggested that elevated GlcCer and decreased S1P levels in neurons are related to neuronal cell death and that elevated LacCer levels induce neurodegeneration by neuroinflammation. In the present study, we found decreased plasma S1P levels and elevated plasma MonCer and LacCer levels in those with neurodegenerative diseases, which is a new finding indicating the importance of abnormal sphingolipid metabolism in neurodegeneration.
Collapse
Affiliation(s)
- Hideki Oizumi
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Yoko Sugimura
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Tomoko Totsune
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Iori Kawasaki
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Saki Ohshiro
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Toru Baba
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Teiko Kimpara
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Hiroaki Sakuma
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Takafumi Hasegawa
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ichiro Kawahata
- Department of Pharmacology, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Atsushi Takeda
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
- Department of Cognitive and Motor Aging, Tohoku University Graduate School of Medicine, Sendai, Japan
- * E-mail:
| |
Collapse
|
20
|
Magadum A. Modified mRNA Therapeutics for Heart Diseases. Int J Mol Sci 2022; 23:ijms232415514. [PMID: 36555159 PMCID: PMC9779737 DOI: 10.3390/ijms232415514] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVD) remain a substantial global health problem and the leading cause of death worldwide. Although many conventional small-molecule treatments are available to support the cardiac function of the patient with CVD, they are not effective as a cure. Among potential targets for gene therapy are severe cardiac and peripheral ischemia, heart failure, vein graft failure, and some forms of dyslipidemias. In the last three decades, multiple gene therapy tools have been used for heart diseases caused by proteins, plasmids, adenovirus, and adeno-associated viruses (AAV), but these remain as unmet clinical needs. These gene therapy methods are ineffective due to poor and uncontrolled gene expression, low stability, immunogenicity, and transfection efficiency. The synthetic modified mRNA (modRNA) presents a novel gene therapy approach which provides a transient, stable, safe, non-immunogenic, controlled mRNA delivery to the heart tissue without any risk of genomic integration, and achieves a therapeutic effect in different organs, including the heart. The mRNA translation starts in minutes, and remains stable for 8-10 days (pulse-like kinetics). The pulse-like expression of modRNA in the heart induces cardiac repair, cardiomyocyte proliferation and survival, and inhibits cardiomyocyte apoptosis post-myocardial infarction (MI). Cell-specific (cardiomyocyte) modRNA translation developments established cell-specific modRNA therapeutics for heart diseases. With these laudable characteristics, combined with its expression kinetics in the heart, modRNA has become an attractive therapeutic for the treatment of CVD. This review discusses new developments in modRNA therapy for heart diseases.
Collapse
Affiliation(s)
- Ajit Magadum
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
21
|
The altered lipidome of hepatocellular carcinoma. Semin Cancer Biol 2022; 86:445-456. [PMID: 35131480 DOI: 10.1016/j.semcancer.2022.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
Abstract
Alterations in metabolic pathways are a hallmark of cancer. A deeper understanding of the contribution of different metabolites to carcinogenesis is thus vitally important to elucidate mechanisms of tumor initiation and progression to inform therapeutic strategies. Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide and its altered metabolic landscape is beginning to unfold with the advancement of technologies. In particular, characterization of the lipidome of human HCCs has accelerated, and together with biochemical analyses, are revealing recurrent patterns of alterations in glycerophospholipid, sphingolipid, cholesterol and bile acid metabolism. These widespread alterations encompass a myriad of lipid species with numerous roles affecting multiple hallmarks of cancer, including aberrant growth signaling, metastasis, evasion of cell death and immunosuppression. In this review, we summarize the current trends and findings of the altered lipidomic landscape of HCC and discuss their potential biological significance for hepatocarcinogenesis.
Collapse
|
22
|
Paranjpe V, Galor A, Grambergs R, Mandal N. The role of sphingolipids in meibomian gland dysfunction and ocular surface inflammation. Ocul Surf 2022; 26:100-110. [PMID: 35973562 PMCID: PMC10259413 DOI: 10.1016/j.jtos.2022.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 11/26/2022]
Abstract
Inflammation occurs in response to tissue injury and invasion of microorganisms and is carried out by the innate and adaptive immune systems, which are regulated by numerous chemokines, cytokines, and lipid mediators. There are four major families of bioactive lipid mediators that play an integral role in inflammation - eicosanoids, sphingolipids (SPL), specialized pro-resolving mediators (SPM), and endocannabinoids. SPL have been historically recognized as important structural components of cellular membranes; their roles as bioactive lipids and inflammatory mediators are recent additions. Major SPL metabolites, including sphingomyelin, ceramide, ceramide 1-phosphate (C1P), sphingosine, sphingosine 1-phosphate (S1P), and their respective enzymes have been studied extensively, primarily in cell-culture and animal models, for their roles in cellular signaling and regulating inflammation and apoptosis. Less focus has been given to the involvement of SPL in eye diseases. As such, the aim of this review was to examine relationships between the SPL family and ocular surface diseases, focusing on their role in disease pathophysiology and discussing the potential of therapeutics that disrupt SPL pathways.
Collapse
Affiliation(s)
- Vikram Paranjpe
- Department of Ophthalmology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA
| | - Anat Galor
- Miami Veterans Administration Medical Center, 1201 NW 16th St, Miami, FL, 33125, USA; Bascom Palmer Eye Institute, University of Miami, 900 NW 17th Street, Miami, FL, 33136, USA.
| | - Richard Grambergs
- Departments of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Sciences Center, Hamilton Eye Institute, 930 Madison Avenue, Memphis, TN, 38163, USA
| | - Nawajes Mandal
- Departments of Ophthalmology, Anatomy and Neurobiology, University of Tennessee Health Sciences Center, Hamilton Eye Institute, 930 Madison Avenue, Memphis, TN, 38163, USA; Memphis VA Medical Center, Memphis, TN, 38104, USA.
| |
Collapse
|
23
|
Rapid Evaporative Ionization Mass Spectrometry-Based Lipidomics for Identification of Canine Mammary Pathology. Int J Mol Sci 2022; 23:ijms231810562. [PMID: 36142485 PMCID: PMC9502565 DOI: 10.3390/ijms231810562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/02/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
The present work proposes the use of a fast analytical platform for the mass spectrometric (MS) profiling of canine mammary tissues in their native form for the building of a predictive statistical model. The latter could be used as a novel diagnostic tool for the real-time identification of different cellular alterations in order to improve tissue resection during veterinary surgery, as previously validated in human oncology. Specifically, Rapid Evaporative Ionization Mass Spectrometry (REIMS) coupled with surgical electrocautery (intelligent knife—iKnife) was used to collect MS data from histologically processed mammary samples, classified into healthy, hyperplastic/dysplastic, mastitis and tumors. Differences in the lipid composition enabled tissue discrimination with an accuracy greater than 90%. The recognition capability of REIMS was tested on unknown mammary samples, and all of them were correctly identified with a correctness score of 98–100%. Triglyceride identification was increased in healthy mammary tissues, while the abundance of phospholipids was observed in altered tissues, reflecting morpho-functional changes in cell membranes, and oxidized species were also tentatively identified as discriminant features. The obtained lipidomic profiles represented unique fingerprints of the samples, suggesting that the iKnife technique is capable of differentiating mammary tissues following chemical changes in cellular metabolism.
Collapse
|
24
|
Feng Y, Kochovski Z, Arenz C, Lu Y, Kneipp J. Structure and Interaction of Ceramide-Containing Liposomes with Gold Nanoparticles as Characterized by SERS and Cryo-EM. THE JOURNAL OF PHYSICAL CHEMISTRY. C, NANOMATERIALS AND INTERFACES 2022; 126:13237-13246. [PMID: 35983312 PMCID: PMC9377338 DOI: 10.1021/acs.jpcc.2c01930] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 06/15/2022] [Indexed: 06/15/2023]
Abstract
Due to the great potential of surface-enhanced Raman scattering (SERS) as local vibrational probe of lipid-nanostructure interaction in lipid bilayers, it is important to characterize these interactions in detail. The interpretation of SERS data of lipids in living cells requires an understanding of how the molecules interact with gold nanostructures and how intermolecular interactions influence the proximity and contact between lipids and nanoparticles. Ceramide, a sphingolipid that acts as important structural component and regulator of biological function, therefore of interest to probing, lacks a phosphocholine head group that is common to many lipids used in liposome models. SERS spectra of liposomes of a mixture of ceramide, phosphatidic acid, and phosphatidylcholine, as well as of pure ceramide and of the phospholipid mixture are reported. Distinct groups of SERS spectra represent varied contributions of the choline, sphingosine, and phosphate head groups and the structures of the acyl chains. Spectral bands related to the state of order of the membrane and moreover to the amide function of the sphingosine head groups indicate that the gold nanoparticles interact with molecules involved in different intermolecular relations. While cryogenic electron microscopy shows the formation of bilayer liposomes in all preparations, pure ceramide was found to also form supramolecular, concentric stacked and densely packed lamellar, nonliposomal structures. That the formation of such supramolecular assemblies supports the intermolecular interactions of ceramide is indicated by the SERS data. The unique spectral features that are assigned to the ceramide-containing lipid model systems here enable an identification of these molecules in biological systems and allow us to obtain information on their structure and interaction by SERS.
Collapse
Affiliation(s)
- Yiqing Feng
- Department
of Chemistry, Humboldt-Universität
zu Berlin, Brook-Taylor-Straße 2, 12489 Berlin, Germany
- Einstein
Center of Catalysis (EC2/BIG-NSE), Technische
Universität Berlin, Marchstraße 6-8, 10587 Berlin, Germany
| | - Zdravko Kochovski
- Department
of Electrochemical Energy Storage, Helmholtz-Zentrum
Berlin für Materialien und Energie, 14109 Berlin, Germany
| | - Christoph Arenz
- Department
of Chemistry, Humboldt-Universität
zu Berlin, Brook-Taylor-Straße 2, 12489 Berlin, Germany
| | - Yan Lu
- Department
of Electrochemical Energy Storage, Helmholtz-Zentrum
Berlin für Materialien und Energie, 14109 Berlin, Germany
- Institute
of Chemistry, University of Potsdam, 14467 Potsdam, Germany
| | - Janina Kneipp
- Department
of Chemistry, Humboldt-Universität
zu Berlin, Brook-Taylor-Straße 2, 12489 Berlin, Germany
| |
Collapse
|
25
|
Custodia A, Romaus-Sanjurjo D, Aramburu-Núñez M, Álvarez-Rafael D, Vázquez-Vázquez L, Camino-Castiñeiras J, Leira Y, Pías-Peleteiro JM, Aldrey JM, Sobrino T, Ouro A. Ceramide/Sphingosine 1-Phosphate Axis as a Key Target for Diagnosis and Treatment in Alzheimer's Disease and Other Neurodegenerative Diseases. Int J Mol Sci 2022; 23:8082. [PMID: 35897658 PMCID: PMC9331765 DOI: 10.3390/ijms23158082] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Alzheimer's disease (AD) is considered the most prevalent neurodegenerative disease and the leading cause of dementia worldwide. Sphingolipids, such as ceramide or sphingosine 1-phosphate, are bioactive molecules implicated in structural and signaling functions. Metabolic dysfunction in the highly conserved pathways to produce sphingolipids may lead to or be a consequence of an underlying disease. Recent studies on transcriptomics and sphingolipidomics have observed alterations in sphingolipid metabolism of both enzymes and metabolites involved in their synthesis in several neurodegenerative diseases, including AD. In this review, we highlight the most relevant findings related to ceramide and neurodegeneration, with a special focus on AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Tomás Sobrino
- Neuro Aging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINCs), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.C.); (D.R.-S.); (M.A.-N.); (D.Á.-R.); (L.V.-V.); (J.C.-C.); (Y.L.); (J.M.P.-P.); (J.M.A.)
| | - Alberto Ouro
- Neuro Aging Laboratory Group (NEURAL), Clinical Neurosciences Research Laboratories (LINCs), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.C.); (D.R.-S.); (M.A.-N.); (D.Á.-R.); (L.V.-V.); (J.C.-C.); (Y.L.); (J.M.P.-P.); (J.M.A.)
| |
Collapse
|
26
|
Changes in the lipidome of water buffalo milk during intramammary infection by non-aureus Staphylococci. Sci Rep 2022; 12:9665. [PMID: 35690599 PMCID: PMC9188581 DOI: 10.1038/s41598-022-13400-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
This study aimed to determine the lipidome of water buffalo milk with intramammary infection (IMI) by non-aureus staphylococci (NAS), also defined as coagulase-negative staphylococci, using an untargeted lipidomic approach. Non-aureus Staphylococci are the most frequently isolated pathogens from dairy water buffalo milk during mastitis. A total of 17 milk samples from quarters affected by NAS-IMI were collected, and the lipidome was determined by liquid chromatography-quadrupole time-of-flight mass spectrometry. The results were compared with the lipidome determined on samples collected from 16 healthy quarters. The study identified 1934 different lipids, which were classified into 15 classes. The abundance of 72 lipids changed in NAS-IMI milk compared to healthy quarters. Significant changes occurred primarily in the class of free fatty acids. The results of this study provided first-time insight into the lipidome of dairy water buffalo milk. Moreover, the present findings provide evidence that NAS-IMI induces changes in water buffalo milk's lipidome.
Collapse
|
27
|
Devarakonda T, Valle Raleigh J, Mauro AG, Lambert JM, Cowart LA, Salloum FN. Chronic treatment with serelaxin mitigates adverse remodeling in a murine model of ischemic heart failure and modulates bioactive sphingolipid signaling. Sci Rep 2022; 12:8897. [PMID: 35614179 PMCID: PMC9132995 DOI: 10.1038/s41598-022-12930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/18/2022] [Indexed: 11/22/2022] Open
Abstract
Relaxin is a pleiotropic hormone demonstrated to confer cardioprotection in animal models of myocardial infarction and ischemic heart failure by modulating inflammation, fibrosis and arrhythmogenesis. Several of these pathways in the ischemic myocardium are intricately tied with the downstream signaling of bioactive sphingolipids, which play an active role during post-infarction remodeling. In this current study, we examined the effects of relaxin on sphingosine 1-phosphate (S1P), and the potential benefits of relaxin treatment on cardiac health in a rodent model of ischemic heart failure. Acute (30 min) and sub-acute (24 h) treatment of primary cardiomyocytes with serelaxin (recombinant human relaxin-2) increased the cardiomyocyte content of S1P. In the rodent model, treatment with relaxin for 28 days following myocardial ischemia by way of permanent left coronary artery occlusion improved survival and cardiac function, reduced fibrosis and apoptosis, and mitigated the expression of several pro-inflammatory and pro-fibrotic markers. The expression of beclin-1 (autophagy marker) was also reduced. The expression of S1P was significantly higher in cardiac tissue and plasma samples extracted from serelaxin-treated mice at day 28. In conclusion, our studies show a significant protection from relaxin in ischemic heart disease, and demonstrate the association between relaxin signaling and S1P generation.
Collapse
Affiliation(s)
- Teja Devarakonda
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Box 980204, Richmond, VA, 23298, USA
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Juan Valle Raleigh
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Box 980204, Richmond, VA, 23298, USA
| | - Adolfo G Mauro
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Box 980204, Richmond, VA, 23298, USA
| | - Johana M Lambert
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Lauren Ashley Cowart
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, USA
- Hunter Holmes McGuire Veterans' Affairs Medical Center, Richmond, VA, USA
| | - Fadi N Salloum
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-070, Box 980204, Richmond, VA, 23298, USA.
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
28
|
Ouro A, Correa-Paz C, Maqueda E, Custodia A, Aramburu-Núñez M, Romaus-Sanjurjo D, Posado-Fernández A, Candamo-Lourido M, Alonso-Alonso ML, Hervella P, Iglesias-Rey R, Castillo J, Campos F, Sobrino T. Involvement of Ceramide Metabolism in Cerebral Ischemia. Front Mol Biosci 2022; 9:864618. [PMID: 35531465 PMCID: PMC9067562 DOI: 10.3389/fmolb.2022.864618] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/11/2022] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke, caused by the interruption of blood flow to the brain and subsequent neuronal death, represents one of the main causes of disability in worldwide. Although reperfusion therapies have shown efficacy in a limited number of patients with acute ischemic stroke, neuroprotective drugs and recovery strategies have been widely assessed, but none of them have been successful in clinical practice. Therefore, the search for new therapeutic approaches is still necessary. Sphingolipids consist of a family of lipidic molecules with both structural and cell signaling functions. Regulation of sphingolipid metabolism is crucial for cell fate and homeostasis in the body. Different works have emphasized the implication of its metabolism in different pathologies, such as diabetes, cancer, neurodegeneration, or atherosclerosis. Other studies have shown its implication in the risk of suffering a stroke and its progression. This review will highlight the implications of sphingolipid metabolism enzymes in acute ischemic stroke.
Collapse
Affiliation(s)
- Alberto Ouro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Clara Correa-Paz
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Elena Maqueda
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Antía Custodia
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Marta Aramburu-Núñez
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Adrián Posado-Fernández
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - María Candamo-Lourido
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Maria Luz Alonso-Alonso
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Francisco Campos
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| |
Collapse
|
29
|
Díaz-Perales A, Escribese MM, Garrido-Arandia M, Obeso D, Izquierdo-Alvarez E, Tome-Amat J, Barber D. The Role of Sphingolipids in Allergic Disorders. FRONTIERS IN ALLERGY 2022; 2:675557. [PMID: 35386967 PMCID: PMC8974723 DOI: 10.3389/falgy.2021.675557] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/10/2021] [Indexed: 12/12/2022] Open
Abstract
Allergy is defined as a complex chronic inflammatory condition in which genetic and environmental factors are implicated. Sphingolipids are involved in multiple biological functions, from cell membrane components to critical signaling molecules. To date, sphingolipids have been studied in different human pathologies such as neurological disorders, cancer, autoimmunity, and infections. Sphingolipid metabolites, in particular, ceramide and sphingosine-1-phosphate (S1P), regulate a diverse range of cellular processes that are important in immunity and inflammation. Moreover, variations in the sphingolipid concentrations have been strongly associated with allergic diseases. This review will focus on the role of sphingolipids in the development of allergic sensitization and allergic inflammation through the activation of immune cells resident in tissues, as well as their role in barrier remodeling and anaphylaxis. The knowledge gained in this emerging field will help to develop new therapeutic options for allergic disorders.
Collapse
Affiliation(s)
- Araceli Díaz-Perales
- Centro de Biotecnología y Genómica de Plantas (CBGP), Universidad Politécnica de Madrid (UPM), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Maria M Escribese
- Basic Medical Sciences Department, Facultad de Medicina, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - María Garrido-Arandia
- Centro de Biotecnología y Genómica de Plantas (CBGP), Universidad Politécnica de Madrid (UPM), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - David Obeso
- Centro de Excelencia en Metabolómica y Bioanálisis (CEMBIO), Facultad de Farmacia, Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Elena Izquierdo-Alvarez
- Basic Medical Sciences Department, Facultad de Medicina, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| | - Jaime Tome-Amat
- Centro de Biotecnología y Genómica de Plantas (CBGP), Universidad Politécnica de Madrid (UPM), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Domingo Barber
- Basic Medical Sciences Department, Facultad de Medicina, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
30
|
Li J, Zheng X, Li X, Yang J, Liu W, Yang L, Liu B. Study on the protective effect and mechanism of Liriodendrin on radiation enteritis in mice. JOURNAL OF RADIATION RESEARCH 2022; 63:213-220. [PMID: 35059715 PMCID: PMC8944324 DOI: 10.1093/jrr/rrab128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/11/2021] [Indexed: 06/01/2023]
Abstract
Patients receiving pelvic or abdominal radiotherapy may experience acute and/or chronic side effects due to gastrointestinal changes. However, effective medicine for treating radiation enteritis has not been found yet. Sargentodoxa cuneata is a famous Chinese medicine used to treat intestinal inflammation, and our research team has found the main biologically active compound through its extraction, which is Liriodendrin. In this study, we found that Liriodendrin can reduce the expression of Cer, Cer1P and S1P in the sphingolipid pathway, thereby reducing the histological damage to the intestinal tract of mice and inhibiting the apoptosis of intestinal tissue cells. In addition, Liriodendrin can reduce the levels of pro-inflammatory cytokines (IL-6 and TNF-α), and it is suggested through flow cytometry that the proportion of neutrophils in the intestinal tissue can decrease due to the existence of Liriodendrin. At the same time, the western blot evaluation revealed that Liriodendrin significantly inhibited the activation of Bcl-2/Bax/Caspase-3 and NF-κB signaling pathways. The results show that Liriodendrin can inhibit intestinal inflammation and intestinal cell apoptosis through the sphingolipid pathway. Therefore, the aforementioned results demonstrated that Liriodendrin may be a promising drug for the treatment of radiation enteritis.
Collapse
Affiliation(s)
| | | | - Xiong Li
- Department of General Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, 610051, China
| | - Jing Yang
- The School of Biological Science and Technology, Chengdu Medical College, 610083, Chengdu, China
| | - Wei Liu
- Department of General Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, 610051, China
| | - Lei Yang
- Tianjin key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Bin Liu
- Corresponding author. Bin Liu, Department of General Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China. E-mail: ; Tel: +86-13980823937; Fax: +86-028-84771387
| |
Collapse
|
31
|
Khan MBN, Iftikhar F, Khan TW, Danish A, Shamsi T, Musharraf SG, Siddiqui AJ. IVS I-5 (G>C) is associated with changes to RBC membrane lipidome in response to Hydroxyurea treatment in β-thalassemia patients. Mol Omics 2022; 18:534-544. [DOI: 10.1039/d2mo00008c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Red Blood Cell’s membrane loses its integrity during hemoglobinopathies like β-thalassemia and sickle cell disease. Various mutations have been associated with β-thalassemia, the most prevalent of which is the IVS-1-5...
Collapse
|
32
|
Roux-Biejat P, Coazzoli M, Marrazzo P, Zecchini S, Di Renzo I, Prata C, Napoli A, Moscheni C, Giovarelli M, Barbalace MC, Catalani E, Bassi MT, De Palma C, Cervia D, Malaguti M, Hrelia S, Clementi E, Perrotta C. Acid Sphingomyelinase Controls Early Phases of Skeletal Muscle Regeneration by Shaping the Macrophage Phenotype. Cells 2021; 10:3028. [PMID: 34831250 PMCID: PMC8616363 DOI: 10.3390/cells10113028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle regeneration is a complex process involving crosstalk between immune cells and myogenic precursor cells, i.e., satellite cells. In this scenario, macrophage recruitment in damaged muscles is a mandatory step for tissue repair since pro-inflammatory M1 macrophages promote the activation of satellite cells, stimulating their proliferation and then, after switching into anti-inflammatory M2 macrophages, they prompt satellite cells' differentiation into myotubes and resolve inflammation. Here, we show that acid sphingomyelinase (ASMase), a key enzyme in sphingolipid metabolism, is activated after skeletal muscle injury induced in vivo by the injection of cardiotoxin. ASMase ablation shortens the early phases of skeletal muscle regeneration without affecting satellite cell behavior. Of interest, ASMase regulates the balance between M1 and M2 macrophages in the injured muscles so that the absence of the enzyme reduces inflammation. The analysis of macrophage populations indicates that these events depend on the altered polarization of M1 macrophages towards an M2 phenotype. Our results unravel a novel role of ASMase in regulating immune response during muscle regeneration/repair and suggest ASMase as a supplemental therapeutic target in conditions of redundant inflammation that impairs muscle recovery.
Collapse
Affiliation(s)
- Paulina Roux-Biejat
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Marco Coazzoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Pasquale Marrazzo
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Ilaria Di Renzo
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy;
| | - Alessandra Napoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Maria Cristina Barbalace
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Maria Teresa Bassi
- Scientific Institute IRCCS “Eugenio Medea”, 23842 Bosisio Parini, Italy;
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, 20129 Milano, Italy;
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
- Scientific Institute IRCCS “Eugenio Medea”, 23842 Bosisio Parini, Italy;
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| |
Collapse
|
33
|
An JN, Kim H, Kim EN, Cho A, Cho Y, Choi YW, Kim JH, Yang SH, Choi BS, Lim CS, Kim YS, Kim KP, Lee JP. Effects of periostin deficiency on kidney aging and lipid metabolism. Aging (Albany NY) 2021; 13:22649-22665. [PMID: 34607314 PMCID: PMC8544301 DOI: 10.18632/aging.203580] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 08/31/2021] [Indexed: 11/25/2022]
Abstract
Periostin plays a crucial role in fibrosis, which is involved in kidney aging. A few studies have shown that lipid metabolism is involved in kidney aging. We investigated the role of periostin in lipid metabolism during kidney aging. Renal function, fibrosis, and inflammatory markers were studied using urine, blood, and tissue samples from wild-type (WT) C57BL/6 mice and Postn-null mice of 2 and 24 months of age. Lipids were quantitatively profiled using liquid chromatography-tandem mass spectrometry in the multiple reaction monitoring mode. Renal function was worse and tubular atrophy/interstitial fibrosis, periostin expression, and inflammatory and fibrotic markers were more severe in aged WT mice than in young WT mice. In aged Postn-null mice, these changes were mitigated. Thirty-five differentially regulated lipids were identified. Phosphatidylcholines, cholesteryl ester, cholesterol, ceramide-1-phosphate, and CCL5 expression were significantly higher in aged WT mice than in aged Postn-null mice. Particularly, linoleic acid, linolenic acid, arachidonic acid, and docosahexaenoic acid differed strongly between the two groups. Lysophosphatidylcholine acyltransferase 2, which converts lysophosphatidylcholine to phosphatidylcholine, was significantly higher in aged WT mice than in aged Postn-null mice. Periostin expression in the kidneys increased with age, and periostin ablation delayed aging. Changes in lipids and their metabolism were found in Postn-null mice. Further research on the precise mechanisms of and relationships between lipid expression and metabolism, kidney aging, and periostin expression is warranted.
Collapse
Affiliation(s)
- Jung Nam An
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, Gyeonggi-do, Korea
| | - Hyoseon Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Korea
| | - Eun Nim Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ara Cho
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Yeongeun Cho
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, Korea
| | - Young Wook Choi
- Department of Urology, College of Medicine, Chung-Ang University, Seoul, Korea
| | - Jin Hyuk Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Seung Hee Yang
- Seoul National University Kidney Research Institute, Seoul, Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Bum Soon Choi
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin, Korea.,Department of Biomedical Science and Technology, Kyung Hee Medical Science Research Institute, Kyung Hee University, Seoul, Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
34
|
Yoon H, Shaw JL, Haigis MC, Greka A. Lipid metabolism in sickness and in health: Emerging regulators of lipotoxicity. Mol Cell 2021; 81:3708-3730. [PMID: 34547235 PMCID: PMC8620413 DOI: 10.1016/j.molcel.2021.08.027] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022]
Abstract
Lipids play crucial roles in signal transduction, contribute to the structural integrity of cellular membranes, and regulate energy metabolism. Questions remain as to which lipid species maintain metabolic homeostasis and which disrupt essential cellular functions, leading to metabolic disorders. Here, we discuss recent advances in understanding lipid metabolism with a focus on catabolism, synthesis, and signaling. Technical advances, including functional genomics, metabolomics, lipidomics, lipid-protein interaction maps, and advances in mass spectrometry, have uncovered new ways to prioritize molecular mechanisms mediating lipid function. By reviewing what is known about the distinct effects of specific lipid species in physiological pathways, we provide a framework for understanding newly identified targets regulating lipid homeostasis with implications for ameliorating metabolic diseases.
Collapse
Affiliation(s)
- Haejin Yoon
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Ludwig Center for Cancer Research at Harvard, Boston, MA 02115, USA
| | - Jillian L Shaw
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Ludwig Center for Cancer Research at Harvard, Boston, MA 02115, USA.
| | - Anna Greka
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
35
|
Prowse N, Hayley S. Microglia and BDNF at the crossroads of stressor related disorders: Towards a unique trophic phenotype. Neurosci Biobehav Rev 2021; 131:135-163. [PMID: 34537262 DOI: 10.1016/j.neubiorev.2021.09.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/16/2022]
Abstract
Stressors ranging from psychogenic/social to neurogenic/injury to systemic/microbial can impact microglial inflammatory processes, but less is known regarding their effects on trophic properties of microglia. Recent studies do suggest that microglia can modulate neuronal plasticity, possibly through brain derived neurotrophic factor (BDNF). This is particularly important given the link between BDNF and neuropsychiatric and neurodegenerative pathology. We posit that certain activated states of microglia play a role in maintaining the delicate balance of BDNF release onto neuronal synapses. This focused review will address how different "activators" influence the expression and release of microglial BDNF and address the question of tropomyosin receptor kinase B (TrkB) expression on microglia. We will then assess sex-based differences in microglial function and BDNF expression, and how microglia are involved in the stress response and related disorders such as depression. Drawing on research from a variety of other disorders, we will highlight challenges and opportunities for modulators that can shift microglia to a "trophic" phenotype with a view to potential therapeutics relevant for stressor-related disorders.
Collapse
Affiliation(s)
- Natalie Prowse
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
36
|
Zhang Y, Zhang X, Lu M, Zou X. Ceramide-1-phosphate and its transfer proteins in eukaryotes. Chem Phys Lipids 2021; 240:105135. [PMID: 34499882 DOI: 10.1016/j.chemphyslip.2021.105135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/31/2021] [Accepted: 09/02/2021] [Indexed: 02/07/2023]
Abstract
Ceramide-1-phosphate (C1P) is a bioactive phosphorylated sphingolipid (SL), produced through the direct phosphorylation of ceramide by ceramide kinase. It plays important roles in regulating cell survival, migration, apoptosis and autophagy and is involved in inflammasome assembly/activation, which can stimulate group IVA cytosolic phospholipase A2α and subsequently increase the levels of arachidonic acid and pro-inflammatory cytokines. Human C1P transfer protein (CPTP) can selectively transport C1P from the Golgi apparatus to specific cellular sites through a non-vesicular mechanism. Human CPTP also affects specific SL levels, thus regulating cell SL homeostasis. In addition, human CPTP plays a crucial role in the regulation of autophagy, inflammation and cell death; thus, human CPTP is closely associated with autophagy and inflammation-related diseases such as cardiovascular and neurodegenerative diseases, and cancers. Therefore, illustrating the functions and mechanisms of human CPTP is important for providing the research foundations for targeted therapy. The key human CPTP residues for C1P recognition and binding are highly conserved in eukaryotic orthologs, while the human CPTP homolog in Arabidopsis (accelerated cell death 11) also exhibits selective inter-membrane transfer of phyto-C1P. These results demonstrate that C1P transporters play fundamental roles in SL metabolism in cells. The present review summarized novel findings of C1P and its TPs in eukaryotes.
Collapse
Affiliation(s)
- Yanqun Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, PR China
| | - Xiangyu Zhang
- Affiliated Stomatology Hospital of Guilin Medical University, Guilin, 541004, PR China
| | - Mengyun Lu
- Affiliated Stomatology Hospital of Guilin Medical University, Guilin, 541004, PR China
| | - Xianqiong Zou
- Affiliated Stomatology Hospital of Guilin Medical University, Guilin, 541004, PR China; College of Biotechnology, Guilin Medical University, Guilin, 541100, PR China.
| |
Collapse
|
37
|
Sindhu S, Leung YH, Arefanian H, Madiraju SRM, Al‐Mulla F, Ahmad R, Prentki M. Neutral sphingomyelinase-2 and cardiometabolic diseases. Obes Rev 2021; 22:e13248. [PMID: 33738905 PMCID: PMC8365731 DOI: 10.1111/obr.13248] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
Sphingolipids, in particular ceramides, play vital role in pathophysiological processes linked to metabolic syndrome, with implications in the development of insulin resistance, pancreatic ß-cell dysfunction, type 2 diabetes, atherosclerosis, inflammation, nonalcoholic steatohepatitis, and cancer. Ceramides are produced by the hydrolysis of sphingomyelin, catalyzed by different sphingomyelinases, including neutral sphingomyelinase 2 (nSMase2), whose dysregulation appears to underlie many of the inflammation-related pathologies. In this review, we discuss the current knowledge on the biochemistry of nSMase2 and ceramide production and its regulation by inflammatory cytokines, with particular reference to cardiometabolic diseases. nSMase2 contribution to pathogenic processes appears to involve cyclical feed-forward interaction with proinflammatory cytokines, such as TNF-α and IL-1ß, which activate nSMase2 and the production of ceramides, that in turn triggers the synthesis and release of inflammatory cytokines. We elaborate these pathogenic interactions at the molecular level and discuss the potential therapeutic benefits of inhibiting nSMase2 against inflammation-driven cardiometabolic diseases.
Collapse
Affiliation(s)
- Sardar Sindhu
- Animal and Imaging core facilityDasman Diabetes InstituteDasmanKuwait
| | - Yat Hei Leung
- Departments of Nutrition, Biochemistry and Molecular MedicineUniversity of MontrealMontréalQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)Montreal Diabetes Research CenterMontréalQuebecCanada
| | - Hossein Arefanian
- Immunology and Microbiology DepartmentDasman Diabetes InstituteDasmanKuwait
| | - S. R. Murthy Madiraju
- Departments of Nutrition, Biochemistry and Molecular MedicineUniversity of MontrealMontréalQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)Montreal Diabetes Research CenterMontréalQuebecCanada
| | - Fahd Al‐Mulla
- Department of Genetics and BioinformaticsDasman Diabetes InstituteDasmanKuwait
| | - Rasheed Ahmad
- Immunology and Microbiology DepartmentDasman Diabetes InstituteDasmanKuwait
| | - Marc Prentki
- Departments of Nutrition, Biochemistry and Molecular MedicineUniversity of MontrealMontréalQuebecCanada
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM)Montreal Diabetes Research CenterMontréalQuebecCanada
| |
Collapse
|
38
|
Ceramide Metabolism Enzymes-Therapeutic Targets against Cancer. ACTA ACUST UNITED AC 2021; 57:medicina57070729. [PMID: 34357010 PMCID: PMC8303233 DOI: 10.3390/medicina57070729] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022]
Abstract
Sphingolipids are both structural molecules that are essential for cell architecture and second messengers that are involved in numerous cell functions. Ceramide is the central hub of sphingolipid metabolism. In addition to being the precursor of complex sphingolipids, ceramides induce cell cycle arrest and promote cell death and inflammation. At least some of the enzymes involved in the regulation of sphingolipid metabolism are altered in carcinogenesis, and some are targets for anticancer drugs. A number of scientific reports have shown how alterations in sphingolipid pools can affect cell proliferation, survival and migration. Determination of sphingolipid levels and the regulation of the enzymes that are implicated in their metabolism is a key factor for developing novel therapeutic strategies or improving conventional therapies. The present review highlights the importance of bioactive sphingolipids and their regulatory enzymes as targets for therapeutic interventions with especial emphasis in carcinogenesis and cancer dissemination.
Collapse
|
39
|
Li L, Ning N, Wei JA, Huang QL, Lu Y, Pang XF, Wu JJ, Zhou JB, Zhou JW, Luo GA, Han L. Metabonomics Study on the Infertility Treated With Zishen Yutai Pills Combined With In Vitro Fertilization-embryo Transfer. Front Pharmacol 2021; 12:686133. [PMID: 34349647 PMCID: PMC8327273 DOI: 10.3389/fphar.2021.686133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/28/2021] [Indexed: 01/10/2023] Open
Abstract
Zishen Yutai Pills (ZYP) is a safe and well quality-controlled TCM preparation with promising effects in many fields of reproduction, including prevention of miscarriage, increase of pregnancy rate during in vitro fertilization-embryo transfer (IVF-ET). The plasma of patients was collected from a clinical trial, namely, “Effect of Traditional Chinese Medicine vs placebo on live births among women undergoing in vitro fertilization, a multi-center randomized controlled trial.” Plasma samples were analyzed with metabonomics method. UPLC-MS technology was used to establish the plasma metabolic fingerprint. Multivariate statistical analysis was applied for comparing the differences of plasma metabolites between ZYP group and placebo group, 44 potential metabolites were screen out and identified. Pathway analysis was conducted with database mining. Compared with placebo, chemicals were found to be significantly down-regulated on HCG trigger day and 14 days after embryo transplantation, including trihexosylceramide (d18:1/26:1), glucosylceramide(d18:1/26:0), TG(22:6/15:0/22:6), TG(22:4/20:4/18:4). Compared with placebo, some chemicals were found to be significantly up-regulated on HCG trigger day and 14 days after embryo transplantation, i.e., PIP3(16:0/16:1), PIP2(18:1/18:1), tauroursodeoxycholic acid, L-asparagine, L-glutamic acid, kynurenic acid, 11-deoxycorticosterone, melatonin glucuronide, hydroxytyrosol. These metabolites were highly enriched in pathways including sphingolipid metabolism, alanine, aspartic acid and glutamic acid metabolism, aminoacyl tRNA biosynthesis, taurine and hypotaurine metabolism. This study revealed metabolic differences between subjects administered with ZYP and placebo. Relating metabolites were identified and pathways were enriched, providing basis on the exploration on the underlying mechanisms of ZYP combined with IVF-ET in the treatment of infertility.
Collapse
Affiliation(s)
- Li Li
- Molecular Biology and Systems Biology Team of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine (The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences), Guangzhou, China
| | - Na Ning
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co. Ltd, Guangzhou, China
| | - Jian-An Wei
- Molecular Biology and Systems Biology Team of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine (The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences), Guangzhou, China
| | - Qiu-Ling Huang
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co. Ltd, Guangzhou, China
| | - Yue Lu
- Molecular Biology and Systems Biology Team of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine (The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences), Guangzhou, China
| | - Xiu-Fei Pang
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co. Ltd, Guangzhou, China
| | - Jing-Jing Wu
- Molecular Biology and Systems Biology Team of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine (The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences), Guangzhou, China
| | - Jie-Bin Zhou
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co. Ltd, Guangzhou, China
| | - Jie-Wen Zhou
- Guangzhou Baiyunshan Zhongyi Pharmaceutical Co. Ltd, Guangzhou, China
| | - Guo-An Luo
- Molecular Biology and Systems Biology Team of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine (The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences), Guangzhou, China
| | - Ling Han
- Molecular Biology and Systems Biology Team of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine (The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences), Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China.,State key laboratory of Dampness Syndrome of Chinese Medicine, The second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
40
|
Custodia A, Aramburu-Núñez M, Correa-Paz C, Posado-Fernández A, Gómez-Larrauri A, Castillo J, Gómez-Muñoz A, Sobrino T, Ouro A. Ceramide Metabolism and Parkinson's Disease-Therapeutic Targets. Biomolecules 2021; 11:945. [PMID: 34202192 PMCID: PMC8301871 DOI: 10.3390/biom11070945] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Ceramide is a bioactive sphingolipid involved in numerous cellular processes. In addition to being the precursor of complex sphingolipids, ceramides can act as second messengers, especially when they are generated at the plasma membrane of cells. Its metabolic dysfunction may lead to or be a consequence of an underlying disease. Recent reports on transcriptomics and electrospray ionization mass spectrometry analysis have demonstrated the variation of specific levels of sphingolipids and enzymes involved in their metabolism in different neurodegenerative diseases. In the present review, we highlight the most relevant discoveries related to ceramide and neurodegeneration, with a special focus on Parkinson's disease.
Collapse
Affiliation(s)
- Antía Custodia
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Marta Aramburu-Núñez
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Clara Correa-Paz
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Adrián Posado-Fernández
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Ana Gómez-Larrauri
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country, P.O. Box 644, 48980 Bilbao, Spain; (A.G.-L.); (A.G.-M.)
- Respiratory Department, Cruces University Hospital, Barakaldo, 48903 Bizkaia, Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Antonio Gómez-Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country, P.O. Box 644, 48980 Bilbao, Spain; (A.G.-L.); (A.G.-M.)
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| | - Alberto Ouro
- Clinical Neurosciences Research Laboratories, Health Research Institute of Santiago de Compostela (IDIS), Travesa da Choupana s/n, 15706 Santiago de Compostela, Spain; (A.C.); (M.A.-N.); (C.C.-P.); (A.P.-F.); (J.C.)
| |
Collapse
|
41
|
Goldschmied JR, Sengupta A, Sharma A, Taylor L, Morales KH, Thase ME, Thase ME, Weljie A, Kayser MS. Treatment of Insomnia with Zaleplon in HIV+ Significantly Improves Sleep and Depression. PSYCHOPHARMACOLOGY BULLETIN 2021; 51:50-64. [PMID: 34421144 PMCID: PMC8374930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
More than 50% of individuals who are HIV positive report insomnia, which can reduce HIV treatment adherence, impair quality of life, and contribute to metabolic dysfunction. Major depressive disorder is also highly comorbid in this population, leading to further impairment. There is evidence that treating insomnia may improve not only sleep, but depression and metabolic function, as well. The present study aimed to examine the effects of pharmacotherapeutic treatment of insomnia on sleep, depression, and metabolic functioning in individuals with HIV. 20 individuals with asymptomatic seropositive HIV and comorbid insomnia and depression were administered zaleplon for 6 weeks. Insomnia severity was assessed using the Insomnia Severity Index and Epworth Sleepiness Scale, and depression severity was assessed using the Quick Inventory of Depression, both prior to treatment and 6 weeks post treatment. Metabolomic changes were assessed using a comprehensive platform measuring ~2000 lipid features and polar metabolites. Linear mixed effects models demonstrated that 6 weeks of treatment with zaleplon significantly improved symptoms of both insomnia and depression. Metabolomic analyses also demonstrated that changes in insomnia severity were associated with significant changes in key branched chain amino acid metabolites. Our results show that improvement in insomnia is associated with reduced depressive symptoms and beneficial metabolomic changes. Additionally, changes in key branched chain amino acid metabolites following treatment may serve as useful biomarkers of treatment response.
Collapse
Affiliation(s)
- Jennifer R Goldschmied
- Goldschmied, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA. Sengupta, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Sharma, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Taylor, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA. Morales, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA; Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA. Thase, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Weljie, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Kayser, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA
| | - Arjun Sengupta
- Goldschmied, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA. Sengupta, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Sharma, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Taylor, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA. Morales, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA; Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA. Thase, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Weljie, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Kayser, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA
| | - Anup Sharma
- Goldschmied, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA. Sengupta, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Sharma, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Taylor, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA. Morales, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA; Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA. Thase, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Weljie, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Kayser, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA
| | - Lynne Taylor
- Goldschmied, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA. Sengupta, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Sharma, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Taylor, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA. Morales, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA; Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA. Thase, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Weljie, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Kayser, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA
| | - Knashawn H Morales
- Goldschmied, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA. Sengupta, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Sharma, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Taylor, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA. Morales, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA; Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA. Thase, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Weljie, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Kayser, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA
| | - Michael E Thase
- Goldschmied, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA. Sengupta, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Sharma, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Taylor, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA. Morales, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA; Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA. Thase, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Weljie, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Kayser, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA
| | - Michael E Thase
- Goldschmied, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA. Sengupta, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Sharma, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Taylor, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA. Morales, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA; Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA. Thase, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Weljie, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Kayser, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA
| | - Aalim Weljie
- Goldschmied, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA. Sengupta, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Sharma, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Taylor, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA. Morales, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA; Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA. Thase, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Weljie, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Kayser, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA
| | - Matthew S Kayser
- Goldschmied, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA. Sengupta, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Sharma, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Taylor, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA. Morales, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA; Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA. Thase, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA. Weljie, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA. Kayser, Department of Psychiatry, University of Pennsylvania, Philadelphia, PA; Chronobiology and Sleep Institute, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
42
|
Tonoyan NM, Chagovets VV, Starodubtseva NL, Tokareva AO, Chingin K, Kozachenko IF, Adamyan LV, Frankevich VE. Alterations in lipid profile upon uterine fibroids and its recurrence. Sci Rep 2021; 11:11447. [PMID: 34075062 PMCID: PMC8169782 DOI: 10.1038/s41598-021-89859-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/30/2021] [Indexed: 11/09/2022] Open
Abstract
Uterine fibroids (UF) is the most common (about 70% cases) type of gynecological disease, with the recurrence rate varying from 11 to 40%. Because UF has no distinct symptomatology and is often asymptomatic, the specific and sensitive diagnosis of UF as well as the assessment for the probability of UF recurrence pose considerable challenge. The aim of this study was to characterize alterations in the lipid profile of tissues associated with the first-time diagnosed UF and recurrent uterine fibroids (RUF) and to explore the potential of mass spectrometry (MS) lipidomics analysis of blood plasma samples for the sensitive and specific determination of UF and RUF with low invasiveness of analysis. MS analysis of lipid levels in the myometrium tissues, fibroids tissues and blood plasma samples was carried out on 66 patients, including 35 patients with first-time diagnosed UF and 31 patients with RUF. The control group consisted of 15 patients who underwent surgical treatment for the intrauterine septum. Fibroids and myometrium tissue samples were analyzed using direct MS approach. Blood plasma samples were analyzed using high performance liquid chromatography hyphened with mass spectrometry (HPLC/MS). MS data were processed by discriminant analysis with projection into latent structures (OPLS-DA). Significant differences were found between the first-time UF, RUF and control group in the levels of lipids involved in the metabolism of glycerophospholipids, sphingolipids, lipids with an ether bond, triglycerides and fatty acids. Significant differences between the control group and the groups with UF and RUF were found in the blood plasma levels of cholesterol esters, triacylglycerols, (lyso) phosphatidylcholines and sphingomyelins. Significant differences between the UF and RUF groups were found in the blood plasma levels of cholesterol esters, phosphotidylcholines, sphingomyelins and triacylglycerols. Diagnostic models based on the selected differential lipids using logistic regression showed sensitivity and specificity of 88% and 86% for the diagnosis of first-time UF and 95% and 79% for RUF, accordingly. This study confirms the involvement of lipids in the pathogenesis of uterine fibroids. A diagnostically significant panel of differential lipid species has been identified for the diagnosis of UF and RUF by low-invasive blood plasma analysis. The developed diagnostic models demonstrated high potential for clinical use and further research in this direction.
Collapse
Affiliation(s)
- Narine M Tonoyan
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, Moscow, 117997, Russian Federation
| | - Vitaliy V Chagovets
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, Moscow, 117997, Russian Federation
| | - Natalia L Starodubtseva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, Moscow, 117997, Russian Federation
- Moscow Institute of Physics and Technology, Moscow Region, 141700, Russian Federation
| | - Alisa O Tokareva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, Moscow, 117997, Russian Federation
- V.L. Talrose Institute for Energy Problems of Chemical Physics, Russia Academy of Sciences, Moscow, 119991, Russian Federation
| | - Konstantin Chingin
- Jiangxi Key Laboratory for Mass Spectrometry and Instrumentation, East China University of Technology, Nanchang, 330013, China
| | - Irena F Kozachenko
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, Moscow, 117997, Russian Federation
| | - Leyla V Adamyan
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, Moscow, 117997, Russian Federation
| | - Vladimir E Frankevich
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov of the Ministry of Healthcare of Russian Federation, Moscow, 117997, Russian Federation.
| |
Collapse
|
43
|
Mahboobnia K, Pirro M, Marini E, Grignani F, Bezsonov EE, Jamialahmadi T, Sahebkar A. PCSK9 and cancer: Rethinking the link. Biomed Pharmacother 2021; 140:111758. [PMID: 34058443 DOI: 10.1016/j.biopha.2021.111758] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cancer is emerging as a major problem globally, as it accounts for the second cause of death despite medical advances. According to epidemiological and basic studies, cholesterol is involved in cancer progression and there are abnormalities in cholesterol metabolism of cancer cells including prostate, breast, and colorectal carcinomas. However, the importance of cholesterol in carcinogenesis and thereby the role of cholesterol homeostasis as a therapeutic target is still a debated area in cancer therapy. Proprotein convertase subtilisin/kexin type-9 (PCSK9), a serine protease, modulates cholesterol metabolism by attachment to the LDL receptor (LDLR) and reducing its recycling by targeting the receptor for lysosomal destruction. Published research has shown that PCSK9 is also involved in degradation of other LDLR family members namely very-low-density-lipoprotein receptor (VLDLR), lipoprotein receptor-related protein 1 (LRP-1), and apolipoprotein E receptor 2 (ApoER2). As a result, this protein represents an interesting therapeutic target for the treatment of hypercholesterolemia. Interestingly, clinical trials on PCSK9-specific monoclonal antibodies have reported promising results with high efficacy in lowering LDL-C and in turn reducing cardiovascular complications. It is important to note that PCSK9 mediates several other pathways apart from its role in lipid homeostasis, including antiviral activity, hepatic regeneration, neuronal apoptosis, and modulation of various signaling pathways. Furthermore, recent literature has illustrated that PCSK9 is closely associated with incidence and progression of several cancers. In a number of studies, PCSK9 siRNA was shown to effectively suppress the proliferation and invasion of the several studied tumor cells. Hence, a novel application of PCSK9 inhibitors/silencers in cancer/metastasis could be considered. However, due to poor data on effectiveness and safety of PCSK9 inhibitors in cancer, the impact of PCSK9 inhibition in these pathological conditions is still unknown. SEARCH METHODS A vast literature search was conducted to find intended studies from 1956 up to 2020, and inclusion criteria were original peer-reviewed publications. PURPOSE OF REVIEW To date, PCSK9 has been scantly investigated in cancer. The question that needs to be discussed is "How does PCSK9 act in cancer pathophysiology and what are the risks or benefits associated to its inhibition?". We reviewed the available publications highlighting the contribution of this proprotein convertase in pathways related to cancer, with focus on the potential implications of its long-term pharmacological inhibition in cancer therapy.
Collapse
Affiliation(s)
- Khadijeh Mahboobnia
- Department of Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Ettore Marini
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Francesco Grignani
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Evgeny E Bezsonov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, 3 Tsyurupa Street, Moscow 117418, Russia; Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow 125315, Russia
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
44
|
Quinville BM, Deschenes NM, Ryckman AE, Walia JS. A Comprehensive Review: Sphingolipid Metabolism and Implications of Disruption in Sphingolipid Homeostasis. Int J Mol Sci 2021; 22:ijms22115793. [PMID: 34071409 PMCID: PMC8198874 DOI: 10.3390/ijms22115793] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
Sphingolipids are a specialized group of lipids essential to the composition of the plasma membrane of many cell types; however, they are primarily localized within the nervous system. The amphipathic properties of sphingolipids enable their participation in a variety of intricate metabolic pathways. Sphingoid bases are the building blocks for all sphingolipid derivatives, comprising a complex class of lipids. The biosynthesis and catabolism of these lipids play an integral role in small- and large-scale body functions, including participation in membrane domains and signalling; cell proliferation, death, migration, and invasiveness; inflammation; and central nervous system development. Recently, sphingolipids have become the focus of several fields of research in the medical and biological sciences, as these bioactive lipids have been identified as potent signalling and messenger molecules. Sphingolipids are now being exploited as therapeutic targets for several pathologies. Here we present a comprehensive review of the structure and metabolism of sphingolipids and their many functional roles within the cell. In addition, we highlight the role of sphingolipids in several pathologies, including inflammatory disease, cystic fibrosis, cancer, Alzheimer’s and Parkinson’s disease, and lysosomal storage disorders.
Collapse
|
45
|
The Role of Ceramide Metabolism and Signaling in the Regulation of Mitophagy and Cancer Therapy. Cancers (Basel) 2021; 13:cancers13102475. [PMID: 34069611 PMCID: PMC8161379 DOI: 10.3390/cancers13102475] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/12/2021] [Accepted: 05/16/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Sphingolipids are membrane-associated lipids that are involved in signal transduction pathways regulating cell death, growth, and migration. In cancer cells, sphingolipids regulate pathways relevant to cancer therapy, such as invasion, metastasis, apoptosis, and lethal mitophagy. Notable sphingolipids include ceramide, a sphingolipid that induces death and lethal mitophagy, and sphingosine-1 phosphate, a sphingolipid that induces survival and chemotherapeutic resistance. These sphingolipids participate in regulating the process of mitophagy, where cells encapsulate damaged mitochondria in double-membrane vesicles (called autophagosomes) for degradation. Lethal mitophagy is an anti-tumorigenic mechanism mediated by ceramide, where cells degrade many mitochondria until the cancer cell dies in an apoptosis-independent manner. Abstract Sphingolipids are bioactive lipids responsible for regulating diverse cellular functions such as proliferation, migration, senescence, and death. These lipids are characterized by a long-chain sphingosine backbone amide-linked to a fatty acyl chain with variable length. The length of the fatty acyl chain is determined by specific ceramide synthases, and this fatty acyl length also determines the sphingolipid’s specialized functions within the cell. One function in particular, the regulation of the selective autophagy of mitochondria, or mitophagy, is closely regulated by ceramide, a key regulatory sphingolipid. Mitophagy alterations have important implications for cancer cell proliferation, response to chemotherapeutics, and mitophagy-mediated cell death. This review will focus on the alterations of ceramide synthases in cancer and sphingolipid regulation of lethal mitophagy, concerning cancer therapy.
Collapse
|
46
|
Xiang H, Jin S, Tan F, Xu Y, Lu Y, Wu T. Physiological functions and therapeutic applications of neutral sphingomyelinase and acid sphingomyelinase. Biomed Pharmacother 2021; 139:111610. [PMID: 33957567 DOI: 10.1016/j.biopha.2021.111610] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 11/15/2022] Open
Abstract
Sphingomyelin (SM) can be converted into ceramide (Cer) by neutral sphingomyelinase (NSM) and acid sphingomyelinase (ASM). Cer is a second messenger of lipids and can regulate cell growth and apoptosis. Increasing evidence shows that NSM and ASM play key roles in many processes, such as apoptosis, immune function and inflammation. Therefore, NSM and ASM have broad prospects in clinical treatments, especially in cancer, cardiovascular diseases (such as atherosclerosis), nervous system diseases (such as Alzheimer's disease), respiratory diseases (such as chronic obstructive pulmonary disease) and the phenotype of dwarfisms in adolescents, playing a complex regulatory role. This review focuses on the physiological functions of NSM and ASM and summarizes their roles in certain diseases and their potential applications in therapy.
Collapse
Affiliation(s)
- Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengjie Jin
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fenglang Tan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifan Xu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
47
|
Gupta P, Taiyab A, Hussain A, Alajmi MF, Islam A, Hassan MI. Targeting the Sphingosine Kinase/Sphingosine-1-Phosphate Signaling Axis in Drug Discovery for Cancer Therapy. Cancers (Basel) 2021; 13:1898. [PMID: 33920887 PMCID: PMC8071327 DOI: 10.3390/cancers13081898] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/11/2021] [Accepted: 04/03/2021] [Indexed: 02/07/2023] Open
Abstract
Sphingolipid metabolites have emerged as critical players in the regulation of various physiological processes. Ceramide and sphingosine induce cell growth arrest and apoptosis, whereas sphingosine-1-phosphate (S1P) promotes cell proliferation and survival. Here, we present an overview of sphingolipid metabolism and the compartmentalization of various sphingolipid metabolites. In addition, the sphingolipid rheostat, a fine metabolic balance between ceramide and S1P, is discussed. Sphingosine kinase (SphK) catalyzes the synthesis of S1P from sphingosine and modulates several cellular processes and is found to be essentially involved in various pathophysiological conditions. The regulation and biological functions of SphK isoforms are discussed. The functions of S1P, along with its receptors, are further highlighted. The up-regulation of SphK is observed in various cancer types and is also linked to radio- and chemoresistance and poor prognosis in cancer patients. Implications of the SphK/S1P signaling axis in human pathologies and its inhibition are discussed in detail. Overall, this review highlights current findings on the SphK/S1P signaling axis from multiple angles, including their functional role, mechanism of activation, involvement in various human malignancies, and inhibitor molecules that may be used in cancer therapy.
Collapse
Affiliation(s)
- Preeti Gupta
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| | - Aaliya Taiyab
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.H.); (M.F.A.)
| | - Mohamed F. Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.H.); (M.F.A.)
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (P.G.); (A.T.); (A.I.)
| |
Collapse
|
48
|
Abstract
Life expectancy, and longevity have been increasing in recent years. However, this is, in most cases, accompanied by age-related diseases. Thus, it became essential to better understand the mechanisms inherent to aging, and to establish biomarkers that characterize this physiological process. Among all biomolecules, lipids appear to be a good target for the study of these biomarkers. In fact, some lipids have already been associated with age-related diseases. With the development of analytical techniques such as Mass Spectrometry, and Nuclear Magnetic Resonance, Lipidomics has been increasingly used to study pathological, and physiological states of an organism. Thus, the study of serum, and plasma lipidome in centenarians, and elderly individuals without age-related diseases can be a useful tool for the identification of aging biomarkers, and to understand physiological aging, and longevity. This review focus on the importance of lipids as biomarkers of aging, and summarize the changes in the lipidome that have been associated with aging, and longevity.
Collapse
|
49
|
Prakash H, Upadhyay D, Bandapalli OR, Jain A, Kleuser B. Host sphingolipids: Perspective immune adjuvant for controlling SARS-CoV-2 infection for managing COVID-19 disease. Prostaglandins Other Lipid Mediat 2020; 152:106504. [PMID: 33147503 PMCID: PMC7605809 DOI: 10.1016/j.prostaglandins.2020.106504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 01/12/2023]
Abstract
That Sphingolipid derivatives are promising drug candidates for the management of novel COVID-19 disease. C-1P based tailoring of Th1 effector immunity for the eradication of infection is a translationally viable approach and deserves immediate attention. That C-1P would promote the killing of infected cells and resolve infection in moderate to severely infected cases. Ceramide derivatives can be exploited as drug candidates for controlling SARS-CoV-2 against novel COVID-19 disease.
Sphingolipids are potent bioactive agents involved in the pathogenesis of various respiratory bacterial infections. To date, several sphingolipid derivatives are known, but S1P (Sphingosine-1-phosphate) and Ceramide are the best-studied sphingolipid derivatives in the context of human diseases. These are membrane-bound lipids that influence host-pathogen interactions. Based on these features, we believe that sphingolipids might control SARS-CoV-2 infection in the host. SARS-CoV-2 utilizes the ACE-II receptor (Angiotensin-converting enzyme II receptor) on epithelial cells for its entry and replication. Activation of the ACE-II receptor is indirectly associated with the activation of S1P Receptor 1 signaling which is associated with IL-6 driven fibrosis. This is expected to promote pathological responses during SARS-CoV-2 infection in COVID-19 cases. Given this, mitigating S1P signaling by application of either S1P Lyase (SPL) or S1P analog (Fingolimod / FTY720) seems to be potential approach for controlling these pathological outcomes. However, due to the immunosuppressive nature of FTY720, it can modulate hyper-inflammatory responses and only provide symptomatic relief, which may not be sufficient for controlling the novel COVID-19 infection. Since Th1 effector immune responses are essential for the clearance of infection, we believe that other sphingolipid derivatives like Cermaide-1 Phosphate with antiviral potential and adjuvant immune potential can potentially control SARS-CoV-2 infection in the host by its ability in enhancing autophagy and antigen presentation by DC to promote T cell response which can be helpful in controlling SARS-CoV-2 infection in novel COVID-19 patients.
Collapse
Affiliation(s)
- Hridayesh Prakash
- Amity Institute of Virology and Immunology, Amity University, Noida, India.
| | - Dilip Upadhyay
- Amity Institute of Virology and Immunology, Amity University, Noida, India
| | | | - Aklank Jain
- Department of Zoology, Central University of Punjab, Bathinda, India
| | - Burkhard Kleuser
- Institute of Nutritional Science, Department of Nutritional Toxicology, University of Potsdam Nuthetal, Germany
| |
Collapse
|
50
|
Kaya I, Sämfors S, Levin M, Borén J, Fletcher JS. Multimodal MALDI Imaging Mass Spectrometry Reveals Spatially Correlated Lipid and Protein Changes in Mouse Heart with Acute Myocardial Infarction. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:2133-2142. [PMID: 32897704 PMCID: PMC7587215 DOI: 10.1021/jasms.0c00245] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Acute myocardial infarction (MI) is a cardiovascular disease that remains a major cause of morbidity and mortality worldwide despite advances in its prevention and treatment. During acute myocardial ischemia, the lack of oxygen switches the cell metabolism to anaerobic respiration, with lactate accumulation, ATP depletion, Na+ and Ca2+ overload, and inhibition of myocardial contractile function, which drastically modifies the lipid, protein, and small metabolite profile in the myocardium. Imaging mass spectrometry (IMS) is a powerful technique to comprehensively elucidate the spatial distribution patterns of lipids, peptides, and proteins in biological tissue sections. In this work, we demonstrate an application of multimodal chemical imaging using matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS), which provided comprehensive molecular information in situ within the same mouse heart tissue sections with myocardial infarction. MALDI-IMS (at 30 μm per pixel) revealed infarct-associated spatial alterations of several lipid species of sphingolipids, glycerophospholipids, lysophospholipids, and cardiolipins along with the acyl carnitines. Further, we performed multimodal MALDI-IMS (IMS3) where dual polarity lipid imaging was combined with subsequent protein MALDI-IMS analysis (at 30 μm per pixel) within the same tissue sections, which revealed accumulations of core histone proteins H4, H2A, and H2B along with post-translational modification products, acetylated H4 and H2A, on the borders of the infarcted region. This methodology allowed us to interpret the lipid and protein molecular pathology of the very same infarcted region in a mouse model of myocardial infarction. Therefore, the presented data highlight the potential of multimodal MALDI imaging mass spectrometry of the same tissue sections as a powerful approach for simultaneous investigation of spatial infarct-associated lipid and protein changes of myocardial infarction.
Collapse
Affiliation(s)
- Ibrahim Kaya
- Department of Psychiatry and Neurochemistry,
Sahlgrenska Academy at the University of Gothenburg, 431 80
Mölndal, Sweden
- Department of Chemistry and Molecular Biology,
University of Gothenburg, 405 30 Gothenburg,
Sweden
| | - Sanna Sämfors
- Department of Chemistry and Molecular Biology,
University of Gothenburg, 405 30 Gothenburg,
Sweden
- Department of Molecular and Clinical Medicine,
Institute of Medicine at University of Gothenburg and Sahlgrenska
University Hospital, 405 30 Gothenburg, Sweden
| | - Malin Levin
- Department of Molecular and Clinical Medicine,
Institute of Medicine at University of Gothenburg and Sahlgrenska
University Hospital, 405 30 Gothenburg, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine,
Institute of Medicine at University of Gothenburg and Sahlgrenska
University Hospital, 405 30 Gothenburg, Sweden
| | - John S. Fletcher
- Department of Chemistry and Molecular Biology,
University of Gothenburg, 405 30 Gothenburg,
Sweden
| |
Collapse
|