1
|
Yan K, Zhang W, Song H, Xu X. Sphingolipid metabolism and regulated cell death in malignant melanoma. Apoptosis 2024; 29:1860-1878. [PMID: 39068623 DOI: 10.1007/s10495-024-02002-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Malignant melanoma (MM) is a highly invasive and therapeutically resistant skin malignancy, posing a significant clinical challenge in its treatment. Programmed cell death plays a crucial role in the occurrence and progression of MM. Sphingolipids (SP), as a class of bioactive lipids, may be associated with many kinds of diseases. SPs regulate various forms of programmed cell death in tumors, including apoptosis, necroptosis, ferroptosis, and more. This review will delve into the mechanisms by which different types of SPs modulate various forms of programmed cell death in MM, such as their regulation of cell membrane permeability and signaling pathways, and how they influence the survival and death fate of MM cells. An in-depth exploration of the role of SPs in programmed cell death in MM aids in unraveling the molecular mechanisms of melanoma development and holds significant importance in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Kexin Yan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, China
| | - Wei Zhang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, China
| | - Hao Song
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, China.
| | - Xiulian Xu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences, Peking Union Medical College, Nanjing, China.
| |
Collapse
|
2
|
Liu B, Zhou J, Jiang B, Tang B, Liu T, Lei P. The role of ACER2 in intestinal sphingolipid metabolism and gastrointestinal cancers. Front Immunol 2024; 15:1511283. [PMID: 39650647 PMCID: PMC11621088 DOI: 10.3389/fimmu.2024.1511283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Sphingolipids, particularly sphingosine-1-phosphate (S1P), are bioactive lipids involved in regulating cellular processes such as proliferation, apoptosis, inflammation, and tumor progression. Alkaline ceramidase 2 (ACER2) plays a critical role in sphingolipid metabolism by catalyzing the hydrolysis of ceramide to sphingosine, which is subsequently converted to S1P. Dysregulation of ACER2 has been implicated in various gastrointestinal cancers, including colorectal cancer, gastric cancer, and hepatocellular carcinoma. ACER2-mediated sphingolipid signaling, particularly through the SphK/S1P pathway, influences cancer development by modulating immune responses, inflammation, and the balance between cell survival and death. This review examines the physiological functions of ACER2, and its role in sphingolipid metabolism, and its contribution to the pathogenesis of gastrointestinal cancers. Understanding the mechanisms by which ACER2 regulates tumor progression and immune modulation may open new avenues for targeted therapies in gastrointestinal malignancies.
Collapse
Affiliation(s)
- Binggang Liu
- Department of Gastrointestinal Surgery, the Central Hospital of Yongzhou, Yongzhou, China
| | | | | | | | | | | |
Collapse
|
3
|
Huang S, Abutaleb K, Mishra S. Glycosphingolipids in Cardiovascular Disease: Insights from Molecular Mechanisms and Heart Failure Models. Biomolecules 2024; 14:1265. [PMID: 39456198 PMCID: PMC11506000 DOI: 10.3390/biom14101265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
This review explores the crucial role of glycosphingolipids (GSLs) in the context of cardiovascular diseases (CVDs), focusing on their biosynthesis, metabolic pathways, and implications for clinical outcomes. GSLs are pivotal in regulating a myriad of cellular functions that are essential for heart health and disease progression. Highlighting findings from both human cohorts and animal models, this review emphasizes the potential of GSLs as biomarkers and therapeutic targets. We advocate for more detailed mechanistic studies to deepen our understanding of GSL functions in cardiovascular health, which could lead to innovative strategies for diagnosis, treatment, and personalized medicine in cardiovascular care.
Collapse
Affiliation(s)
- Sarah Huang
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Karima Abutaleb
- Department of Surgery, Virginia Tech Carilion School of Medicine, Roanoke, VA 24061, USA
| | - Sumita Mishra
- Department of Surgery, Virginia Tech Carilion School of Medicine, Roanoke, VA 24061, USA
- Center for Exercise Medicine Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24061, USA
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA 24061, USA
- Department of Human Nutrition, Foods, and Exercise, College of Life Sciences, Virginia Tech, Roanoke, VA 24061, USA
| |
Collapse
|
4
|
Li H, Zhang L, Yang F, Feng X, Fu R, Zhao R, Li X, Li H. Lipid-lowering drugs affect lung cancer risk via sphingolipid metabolism: a drug-target Mendelian randomization study. Front Genet 2023; 14:1269291. [PMID: 38034491 PMCID: PMC10687161 DOI: 10.3389/fgene.2023.1269291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
Background: The causal relationship between lipid-lowering drug (LLD) use and lung cancer risk is controversial, and the role of sphingolipid metabolism in this effect remains unclear. Methods: Genome-wide association study data on low-density lipoprotein (LDL), apolipoprotein B (ApoB), and triglycerides (TG) were used to develop genetic instrumental variables (IVs) for LLDs. Two-step Mendelian randomization analyses were performed to examine the causal relationship between LLDs and lung cancer risk. The effects of ceramide, sphingosine-1-phosphate (S1P), and ceramidases on lung cancer risk were explored, and the proportions of the effects of LLDs on lung cancer risk mediated by sphingolipid metabolism were calculated. Results: APOB inhibition decreased the lung cancer risk in ever-smokers via ApoB (odds ratio [OR] 0.81, 95% confidence interval [CI] 0.70-0.92, p = 0.010), LDL (OR 0.82, 95% CI 0.71-0.96, p = 0.040), and TG (OR 0.63, 95% CI 0.46-0.83, p = 0.015) reduction by 1 standard deviation (SD), decreased small-cell lung cancer (SCLC) risk via LDL reduction by 1 SD (OR 0.71, 95% CI 0.56-0.90, p = 0.016), and decreased the plasma ceramide level and increased the neutral ceramidase level. APOC3 inhibition decreased the lung adenocarcinoma (LUAD) risk (OR 0.60, 95% CI 0.43-0.84, p = 0.039) but increased SCLC risk (OR 2.18, 95% CI 1.17-4.09, p = 0.029) via ApoB reduction by 1 SD. HMGCR inhibition increased SCLC risk via ApoB reduction by 1 SD (OR 3.04, 95% CI 1.38-6.70, p = 0.014). The LPL agonist decreased SCLC risk via ApoB (OR 0.20, 95% CI 0.07-0.58, p = 0.012) and TG reduction (OR 0.58, 95% CI 0.43-0.77, p = 0.003) while increased the plasma S1P level. PCSK9 inhibition decreased the ceramide level. Neutral ceramidase mediated 8.1% and 9.5% of the reduced lung cancer risk in ever-smokers via ApoB and TG reduction by APOB inhibition, respectively, and mediated 8.7% of the reduced LUAD risk via ApoB reduction by APOC3 inhibition. Conclusion: We elucidated the intricate interplay between LLDs, sphingolipid metabolites, and lung cancer risk. Associations of APOB, APOC3, and HMGCR inhibition and LPL agonist with distinct lung cancer risks underscore the multifaceted nature of these relationships. The observed mediation effects highlight the considerable influence of neutral ceramidase on the lung cancer risk reduction achieved by APOB and APOC3 inhibition.
Collapse
Affiliation(s)
- Honglin Li
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lei Zhang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Feiran Yang
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaoteng Feng
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Fu
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ruohan Zhao
- Department of Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiurong Li
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Huijie Li
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
5
|
Le Guillou S, Ciobotaru C, Laubier J, Castille J, Aujean E, Hue-Beauvais C, Cherbuy C, Liuu S, Henry C, David A, Jaffrezic F, Laloë D, Charlier M, Alexandre-Gouabau MC, Le Provost F. Specific Milk Composition of miR-30b Transgenic Mice Associated with Early Duodenum Maturation in Offspring with Lasting Consequences for Growth. J Nutr 2023; 153:2808-2826. [PMID: 37543213 DOI: 10.1016/j.tjnut.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 07/18/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023] Open
Abstract
BACKGROUND Milk composition is complex and includes numerous components essential for offspring growth and development. In addition to the high abundance of miR-30b microRNA, milk produced by the transgenic mouse model of miR-30b-mammary deregulation displays a significantly altered fatty acid profile. Moreover, wild-type adopted pups fed miR-30b milk present an early growth defect. OBJECTIVE This study aimed to investigate the consequences of miR-30b milk feeding on the duodenal development of wild-type neonates, a prime target of suckled milk, along with comprehensive milk phenotyping. METHODS The duodenums of wild-type pups fed miR-30b milk were extensively characterized at postnatal day (PND)-5, PND-6, and PND-15 using histological, transcriptomic, proteomic, and duodenal permeability analyses and compared with those of pups fed wild-type milk. Milk of miR-30b foster dams collected at mid-lactation was extensively analyzed using proteomic, metabolomic, and lipidomic approaches and hormonal immunoassays. RESULTS At PND-5, wild-type pups fed miR-30b milk showed maturation of their duodenum with 1.5-fold (P < 0.05) and 1.3-fold (P < 0.10) increased expression of Claudin-3 and Claudin-4, respectively, and changes in 8 duodenal proteins (P < 0.10), with an earlier reduction in paracellular and transcellular permeability (183 ng/mL fluorescein sulfonic acid [FSA] and 12 ng/mL horseradish peroxidase [HRP], respectively, compared with 5700 ng/mL FSA and 90 ng/mL HRP in wild-type; P < 0.001). Compared with wild-type milk, miR-30b milk displayed an increase in total lipid (219 g/L compared with 151 g/L; P < 0.05), ceramide (17.6 μM compared with 6.9 μM; P < 0.05), and sphingomyelin concentrations (163.7 μM compared with 76.3 μM; P < 0.05); overexpression of 9 proteins involved in the gut barrier (P < 0.1); and higher insulin and leptin concentrations (1.88 ng/mL and 2.04 ng/mL, respectively, compared with 0.79 ng/mL and 1.06 ng/mL; P < 0.01). CONCLUSIONS miR-30b milk displays significant changes in bioactive components associated with neonatal duodenal integrity and maturation, which could be involved in the earlier intestinal closure phenotype of the wild-type pups associated with a lower growth rate.
Collapse
Affiliation(s)
| | - Céline Ciobotaru
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Johann Laubier
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Johan Castille
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Etienne Aujean
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Cathy Hue-Beauvais
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Claire Cherbuy
- Université Paris-Saclay, INRAE, MICALIS Institute, Jouy-en-Josas, France
| | - Sophie Liuu
- Université Paris-Saclay, INRAE, AgroParisTech, MICALIS Institute, PAPPSO, Jouy-en-Josas, France
| | - Céline Henry
- Université Paris-Saclay, INRAE, AgroParisTech, MICALIS Institute, PAPPSO, Jouy-en-Josas, France
| | - Agnès David
- Nantes Université, CRNH-OUEST, INRAE, UMR 1280, PhAN, Nantes, France
| | - Florence Jaffrezic
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Denis Laloë
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | - Madia Charlier
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | | | | |
Collapse
|
6
|
Yan T, Yang H, Xu C, Liu J, Meng Y, Jiang Q, Li J, Kang G, Zhou L, Xiao S, Xue Y, Xu J, Chen X, Che F. Inhibition of hyaluronic acid degradation pathway suppresses glioma progression by inducing apoptosis and cell cycle arrest. Cancer Cell Int 2023; 23:163. [PMID: 37568202 PMCID: PMC10422813 DOI: 10.1186/s12935-023-02998-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Abnormal hyaluronic acid (HA) metabolism is a major factor in tumor progression, and the metabolic regulation of HA mainly includes HA biosynthesis and catabolism. In glioma, abnormal HA biosynthesis is intimately involved in glioma malignant biological properties and the formation of immunosuppressive microenvironment; however, the role of abnormal HA catabolism in glioma remains unclear. METHODS HA catabolism is dependent on hyaluronidase. In TCGA and GEPIA databases, we found that among the 6 human hyaluronidases (HYAL1, HYAL2, HYAL3, HYAL4, HYALP1, SPAM1), only HYAL2 expression was highest in glioma. Next, TCGA and CGGA database were further used to explore the correlation of HYAL2 expression with glioma prognosis. Then, the mRNA expression and protein level of HYAL2 was determined by qRT-PCR, Western blot and Immunohistochemical staining in glioma cells and glioma tissues, respectively. The MTT, EdU and Colony formation assay were used to measure the effect of HYAL2 knockdown on glioma. The GSEA enrichment analysis was performed to explore the potential pathway regulated by HYAL2 in glioma, in addition, the HYAL2-regulated signaling pathways were detected by flow cytometry and Western blot. Finally, small molecule compounds targeting HYAL2 in glioma were screened by Cmap analysis. RESULTS In the present study, we confirmed that Hyaluronidase 2 (HYAL2) is abnormally overexpressed in glioma. Moreover, we found that HYAL2 overexpression is associated with multiple glioma clinical traits and acts as a key indicator for glioma prognosis. Targeting HYAL2 could inhibit glioma progression by inducing glioma cell apoptosis and cell cycle arrest. CONCLUSION Collectively, these observations suggest that HYAL2 overexpression could promote glioma progression. Thus, treatments that disrupt HA catabolism by altering HYAL2 expression may serve as effective strategies for glioma treatment.
Collapse
Affiliation(s)
- Tao Yan
- Central Laboratory, Linyi People's Hospital, Guangzhou University of Chinese Medicine, Linyi, 276000, Shandong Province, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - He Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Caixia Xu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Junsi Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yun Meng
- Central Laboratory, Linyi People's Hospital, Guangzhou University of Chinese Medicine, Linyi, 276000, Shandong Province, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Qing Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Jinxing Li
- Department of Neurosurgery, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Guiqiong Kang
- Central Laboratory, Linyi People's Hospital, Guangzhou University of Chinese Medicine, Linyi, 276000, Shandong Province, China
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Liangjian Zhou
- Scientific Research Management Office, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Shuai Xiao
- Scientific Research Management Office, Linyi People's Hospital, Linyi, 276000, Shandong Province, China
| | - Yanpeng Xue
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Jiayi Xu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Xin Chen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
- Key Colleges and Universities Laboratory of Neurosurgery in Heilongjiang Province, Harbin, 150001, Heilongjiang Province, China.
- Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
| | - Fengyuan Che
- Central Laboratory, Linyi People's Hospital, Guangzhou University of Chinese Medicine, Linyi, 276000, Shandong Province, China.
- Linyi Key Laboratory of Neurophysiology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China.
- Department of Neurology, Linyi People's Hospital, Linyi, 276000, Shandong Province, China.
| |
Collapse
|
7
|
Nie M, Liu T, Qiu X, Yang J, Liu J, Ren J, Zhou B. Regulation mechanism of lipids for extracellular yellow pigments production by Monascus purpureus BWY-5. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12654-6. [PMID: 37405437 DOI: 10.1007/s00253-023-12654-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/14/2023] [Accepted: 06/18/2023] [Indexed: 07/06/2023]
Abstract
The biosynthesis and secretion of Monascus pigments are closely related to the integrity of the cell membrane, which determines the composition of lipids and its content in cell membrane. The present study aimed to thoroughly describe the changes of lipid profiling in Monascus purpureus BWY-5, which was screened by carbon ion beam irradiation (12C6+) to almost single yield extracellular Monascus yellow pigments (extra-MYPs), by absolute quantitative lipidomics and tandem mass tags (TMT) based quantitative proteomic. 12C6+ irradiation caused non-lipid oxidation damage to Monascus cell membrane, leading to an imbalance in cell membrane lipid homeostasis. This imbalance was attributed to significant changes not only in the composition but also in the content of lipids in Monascus, especially the inhibition of glycerophospholipid biosynthesis. Integrity of plasma membrane was maintained by the increased production of ergosterol, monogalactosylmonoacylglycerol (MGMG) and sulfoquinovosylmonoacylglycerol (SQMG), while mitochondrial membrane homeostasis was maintained by the increase of cardiolipin production. The growth and extra-MYPs production of Monascus BWY-5 have been regulated by the promotion of sphingolipids (ceramide and sulfatide) biosynthesis. Simultaneous, energy homeostasis may be achieved by increase of TG synthesis and Ca2+/Mg2+-ATPase activity. These finding suggest ergosterol, cardiolipin, sphingolipids, MGMG and SQMG play a key facilitating role in cytomembrane lipid homeostasis maintaining for Monascus purpureus BWY-5, and then it is closely related to cell growth and extra-MYPs production. KEY POINTS: 1. Energy homeostasis in Monascus purpureus BWY-5 was achieved by increase of TG synthesis and Ca2+/Mg2+-ATPase activity. 2. Integrity of plasma membrane in Monascus purpureus BWY-5 was maintained by the increased production of ergosterol. 3. Mitochondrial membrane homeostasis in Monascus purpureus BWY-5 was maintaed by the increase of cardiolipin synthesis.
Collapse
Affiliation(s)
- Moyu Nie
- Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Changsha, 410004, Hunan, China
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China
| | - Tao Liu
- Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Changsha, 410004, Hunan, China
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China
| | - Xunhan Qiu
- Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Changsha, 410004, Hunan, China
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China
| | - Jingjing Yang
- Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Changsha, 410004, Hunan, China
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China
| | - Jun Liu
- Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Changsha, 410004, Hunan, China
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China
| | - Jiali Ren
- Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Changsha, 410004, Hunan, China
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China
| | - Bo Zhou
- Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Changsha, 410004, Hunan, China.
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, Hunan, China.
| |
Collapse
|
8
|
Liu JX, Liu M, Yu GZ, Zhao QQ, Wang JL, Sun YH, Koda S, Zhang B, Yu Q, Yan C, Tang RX, Jiang ZH, Zheng KY. Clonorchis sinensis infection induces hepatobiliary injury via disturbing sphingolipid metabolism and activating sphingosine 1-phosphate receptor 2. Front Cell Infect Microbiol 2022; 12:1011378. [PMID: 36339341 PMCID: PMC9627039 DOI: 10.3389/fcimb.2022.1011378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/20/2022] [Indexed: 01/31/2024] Open
Abstract
Clonorchis sinensis (C. sinensis) infection induces severe hepatobiliary injuries, which can cause inflammation, periductal fibrosis, and even cholangiocarcinoma. Sphingolipid metabolic pathways responsible for the generation of sphingosine-1-phosphate (S1P) and its receptor S1P receptors (S1PRs) have been implicated in many liver-related diseases. However, the role of S1PRs in C. sinensis-mediated biliary epithelial cells (BECs) proliferation and hepatobiliary injury has not been elucidated. In the present study, we found that C. sinensis infection resulted in alteration of bioactive lipids and sphingolipid metabolic pathways in mice liver. Furthermore, S1PR2 was predominantly activated among these S1PRs in BECs both in vivo and in vitro. Using JTE-013, a specific antagonist of S1PR2, we found that the hepatobiliary pathological injuries, inflammation, bile duct hyperplasia, and periductal fibrosis can be significantly inhibited in C. sinensis-infected mice. In addition, both C. sinensis excretory-secretory products (CsESPs)- and S1P-induced activation of AKT and ERK1/2 were inhibited by JTE-013 in BECs. Therefore, the sphingolipid metabolism pathway and S1PR2 play an important role, and may serve as potential therapeutic targets in hepatobiliary injury caused by C. sinensis-infection.
Collapse
Affiliation(s)
- Ji-Xin Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- Department of Clinical Pathogen Biology, Qiqihaer Medical University, Qiqihaer, China
| | - Man Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Guo-Zhi Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Qian-Qian Zhao
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Jian-Ling Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Yan-Hong Sun
- Department of Pathogen Biology, Qiqihaer Medical University, Qiqihaer, China
| | - Stephane Koda
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Beibei Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Qian Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Ren-Xian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Zhi-Hua Jiang
- Institute of Parasitic Disease Control and Prevention, Guangxi Key Laboratory for the Prevention and Control of Viral Hepatitis, Guangxi Zhuang Autonomous Region Center for Disease Control and Prevention, Nanning, China
| | - Kui-Yang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Janneh AH, Ogretmen B. Targeting Sphingolipid Metabolism as a Therapeutic Strategy in Cancer Treatment. Cancers (Basel) 2022; 14:2183. [PMID: 35565311 PMCID: PMC9104917 DOI: 10.3390/cancers14092183] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
Sphingolipids are bioactive molecules that have key roles in regulating tumor cell death and survival through, in part, the functional roles of ceramide accumulation and sphingosine-1-phosphate (S1P) production, respectively. Mechanistic studies using cell lines, mouse models, or human tumors have revealed crucial roles of sphingolipid metabolic signaling in regulating tumor progression in response to anticancer therapy. Specifically, studies to understand ceramide and S1P production pathways with their downstream targets have provided novel therapeutic strategies for cancer treatment. In this review, we present recent evidence of the critical roles of sphingolipids and their metabolic enzymes in regulating tumor progression via mechanisms involving cell death or survival. The roles of S1P in enabling tumor growth/metastasis and conferring cancer resistance to existing therapeutics are also highlighted. Additionally, using the publicly available transcriptomic database, we assess the prognostic values of key sphingolipid enzymes on the overall survival of patients with different malignancies and present studies that highlight their clinical implications for anticancer treatment.
Collapse
Affiliation(s)
| | - Besim Ogretmen
- Hollings Cancer Center, Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
12
|
Simoes M, Saleh A, Choi YM, Airola MV, Haley JD, Coant N. Measurement of neutral ceramidase activity in vitro and in vivo. Anal Biochem 2022; 643:114577. [PMID: 35134389 PMCID: PMC11787886 DOI: 10.1016/j.ab.2022.114577] [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: 12/01/2021] [Revised: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 11/01/2022]
Abstract
Neutral ceramidase is a hydrolase of ceramide that has been implicated in multiple biologic processes, including inflammation and oncogenesis. Ceramides and other sphingolipids, belong to a family of N-acyl linked lipids that are biologically active in signaling, despite their limited structural functions. Ceramides are generally pro-apoptotic, while sphingosine and sphingosine-1-phosphate (S1P) exert proliferative and pro-oncogenic effects. Ceramidases are important regulators of ceramide levels that hydrolyze ceramide to sphingosine. Thus, ceramidase inhibition significantly increases the quantities of ceramide and its associated signaling. To better understand the function of ceramide, biochemical and cellular assays for enzymatic activity were developed and validated to identify inhibitors of human neutral ceramidase (nCDase). Here we review the measurement of nCDase activity both in vitro and in vivo.
Collapse
Affiliation(s)
- Michael Simoes
- Department of Pathology and Stony Brook Cancer Center, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794, USA
| | - Amalia Saleh
- Department of Pathology and Stony Brook Cancer Center, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794, USA
| | - Yong-Mi Choi
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Michael V Airola
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - John D Haley
- Department of Pathology and Stony Brook Cancer Center, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794, USA
| | - Nicolas Coant
- Department of Pathology and Stony Brook Cancer Center, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, 11794, USA.
| |
Collapse
|
13
|
Sears SM, Dupre TV, Shah PP, Davis DL, Doll MA, Sharp CN, Vega AA, Megyesi J, Beverly LJ, Snider AJ, Obeid LM, Hannun YA, Siskind LJ. Neutral ceramidase deficiency protects against cisplatin-induced acute kidney injury. J Lipid Res 2022; 63:100179. [PMID: 35151662 PMCID: PMC8953688 DOI: 10.1016/j.jlr.2022.100179] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Cisplatin is a commonly used chemotherapeutic for the treatment of many solid organ cancers; however, its effectiveness is limited by the development of acute kidney injury (AKI) in 30% of patients. AKI is driven by proximal tubule cell death, leading to rapid decline in renal function. It has previously been shown that sphingolipid metabolism plays a role in regulating many of the biological processes involved in cisplatin-induced AKI. For example, neutral ceramidase (nCDase) is an enzyme responsible for converting ceramide into sphingosine, which is then phosphorylated to become sphingosine-1-phosphate, and our lab previously demonstrated that nCDase knockout (nCDase-/-) in mouse embryonic fibroblasts led to resistance to nutrient and energy deprivation-induced cell death via upregulation of autophagic flux. In this study, we further characterized the role of nCDase in AKI by demonstrating that nCDase-/- mice are resistant to cisplatin-induced AKI. nCDase-/- mice display improved kidney function, reduced injury and structural damage, lower rates of apoptosis, and less ER stress compared to wild-type mice following cisplatin treatment. Although the mechanism of protection is still unknown, we propose that it could be mediated by increased autophagy, as chloroquine treatment resensitized nCDase-/- mice to AKI development. Taken together, we conclude that nCDase may represent a novel target to prevent cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Sophia M Sears
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA
| | - Tess V Dupre
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA
| | - Parag P Shah
- Department of Medicine, University of Louisville, Louisville, KY, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Deanna L Davis
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA
| | - Mark A Doll
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA
| | - Cierra N Sharp
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA
| | - Alexis A Vega
- Department of Biochemistry & Molecular Genetics, University of Louisville, Louisville, KY, USA
| | - Judit Megyesi
- Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences and Central Arkansas, Veterans Healthcare System, Little Rock, AR, USA
| | - Levi J Beverly
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA; Department of Medicine, University of Louisville, Louisville, KY, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Ashley J Snider
- Department of Nutritional Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, USA
| | - Lina M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA; Northport Veteran Affairs Medical Center, Northport, NY, USA
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA; Northport Veteran Affairs Medical Center, Northport, NY, USA
| | - Leah J Siskind
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
14
|
Sphingolipid control of cognitive functions in health and disease. Prog Lipid Res 2022; 86:101162. [DOI: 10.1016/j.plipres.2022.101162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 12/14/2022]
|
15
|
Taniai T, Shirai Y, Shimada Y, Hamura R, Yanagaki M, Takada N, Horiuchi T, Haruki K, Furukawa K, Uwagawa T, Tsuboi K, Okamoto Y, Shimada S, Tanaka S, Ohashi T, Ikegami T. Inhibition of acid ceramidase elicits mitochondrial dysfunction and oxidative stress in pancreatic cancer cells. Cancer Sci 2021; 112:4570-4579. [PMID: 34459070 PMCID: PMC8586682 DOI: 10.1111/cas.15123] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/05/2021] [Accepted: 08/11/2021] [Indexed: 01/18/2023] Open
Abstract
Although the inhibition of acid ceramidase (AC) is known to induce antitumor effects in various cancers, there are few reports in pancreatic cancer, and the underlying mechanisms remain unclear. Moreover, there is currently no safe administration method of AC inhibitor. Here the effects of gene therapy using siRNA and shRNA for AC inhibition with its mechanisms for pancreatic cancer were investigated. The inhibition of AC by siRNA and shRNA using an adeno-associated virus 8 (AAV8) vector had antiproliferative effects by inducing apoptosis in pancreatic cancer cells and xenograft mouse model. Acid ceramidase inhibition elicits mitochondrial dysfunction, reactive oxygen species accumulation, and manganese superoxide dismutase suppression, resulting in apoptosis of pancreatic cancer cells accompanied by ceramide accumulation. These results elucidated the mechanisms underlying the antitumor effect of AC inhibition in pancreatic cancer cells and suggest the potential of the AAV8 vector to inhibit AC as a therapeutic strategy.
Collapse
Affiliation(s)
- Tomohiko Taniai
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
- Division of Gene TherapyResearch Center for Medical ScienceThe Jikei University School of MedicineTokyoJapan
| | - Yoshihiro Shirai
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
- Division of Gene TherapyResearch Center for Medical ScienceThe Jikei University School of MedicineTokyoJapan
| | - Yohta Shimada
- Division of Gene TherapyResearch Center for Medical ScienceThe Jikei University School of MedicineTokyoJapan
| | - Ryoga Hamura
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
- Division of Gene TherapyResearch Center for Medical ScienceThe Jikei University School of MedicineTokyoJapan
| | - Mitsuru Yanagaki
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
- Division of Gene TherapyResearch Center for Medical ScienceThe Jikei University School of MedicineTokyoJapan
| | - Naoki Takada
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
- Division of Gene TherapyResearch Center for Medical ScienceThe Jikei University School of MedicineTokyoJapan
| | - Takashi Horiuchi
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
- Division of Gene TherapyResearch Center for Medical ScienceThe Jikei University School of MedicineTokyoJapan
| | - Koichiro Haruki
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
- Division of Gene TherapyResearch Center for Medical ScienceThe Jikei University School of MedicineTokyoJapan
| | - Kenei Furukawa
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
- Division of Gene TherapyResearch Center for Medical ScienceThe Jikei University School of MedicineTokyoJapan
| | - Tadashi Uwagawa
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
| | - Kazuhito Tsuboi
- Department of PharmacologyKawasaki Medical SchoolKurashikiJapan
| | - Yasuo Okamoto
- Department of PharmacologyKawasaki Medical SchoolKurashikiJapan
| | - Shu Shimada
- Department of Molecular Oncology Graduate School of MedicineTokyo Medical and Dental UniversityTokyoJapan
| | - Shinji Tanaka
- Department of Molecular Oncology Graduate School of MedicineTokyo Medical and Dental UniversityTokyoJapan
| | - Toya Ohashi
- Division of Gene TherapyResearch Center for Medical ScienceThe Jikei University School of MedicineTokyoJapan
| | - Toru Ikegami
- Department of SurgeryThe Jikei University School of MedicineTokyoJapan
| |
Collapse
|
16
|
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
|
17
|
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: 35] [Impact Index Per Article: 8.8] [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
|
18
|
Zhu H, Klement JD, Lu C, Redd PS, Yang D, Smith AD, Poschel DB, Zou J, Liu D, Wang PG, Ostrov D, Coant N, Hannun YA, Colby AH, Grinstaff MW, Liu K. Asah2 Represses the p53-Hmox1 Axis to Protect Myeloid-Derived Suppressor Cells from Ferroptosis. THE JOURNAL OF IMMUNOLOGY 2021; 206:1395-1404. [PMID: 33547170 DOI: 10.4049/jimmunol.2000500] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 01/12/2021] [Indexed: 02/07/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are immune suppressive cells that massively accumulate under pathological conditions to suppress T cell immune response. Dysregulated cell death contributes to MDSC accumulation, but the molecular mechanism underlying this cell death dysregulation is not fully understood. In this study, we report that neutral ceramidase (N-acylsphingosine amidohydrolase [ASAH2]) is highly expressed in tumor-infiltrating MDSCs in colon carcinoma and acts as an MDSC survival factor. To target ASAH2, we performed molecular docking based on human ASAH2 protein structure. Enzymatic inhibition analysis of identified hits determined NC06 as an ASAH2 inhibitor. Chemical and nuclear magnetic resonance analysis determined NC06 as 7-chloro-2-(3-chloroanilino)pyrano[3,4-e][1,3]oxazine-4,5-dione. NC06 inhibits ceramidase activity with an IC50 of 10.16-25.91 μM for human ASAH2 and 18.6-30.2 μM for mouse Asah2 proteins. NC06 induces MDSC death in a dose-dependent manner, and inhibition of ferroptosis decreased NC06-induced MDSC death. NC06 increases glutathione synthesis and decreases lipid reactive oxygen species to suppress ferroptosis in MDSCs. Gene expression profiling identified the p53 pathway as the Asah2 target in MDSCs. Inhibition of Asah2 increased p53 protein stability to upregulate Hmox1 expression to suppress lipid reactive oxygen species production to suppress ferroptosis in MDSCs. NC06 therapy increases MDSC death and reduces MDSC accumulation in tumor-bearing mice, resulting in increased activation of tumor-infiltrating CTLs and suppression of tumor growth in vivo. Our data indicate that ASAH2 protects MDSCs from ferroptosis through destabilizing p53 protein to suppress the p53 pathway in MDSCs in the tumor microenvironment. Targeting ASAH2 with NC06 to induce MDSC ferroptosis is potentially an effective therapy to suppress MDSC accumulation in cancer immunotherapy.
Collapse
Affiliation(s)
- Huabin Zhu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912.,Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30904
| | - John D Klement
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912.,Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30904
| | - Chunwan Lu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912.,Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30904
| | - Priscilla S Redd
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912.,Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30904
| | - Dafeng Yang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912.,Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30904
| | - Alyssa D Smith
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912.,Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30904
| | - Dakota B Poschel
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912.,Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30904
| | - Juan Zou
- Department of Chemistry and Physics, Augusta University, Augusta, GA 30912
| | - Ding Liu
- Department of Chemistry, Georgia State University, Atlanta, GA 30303
| | - Peng George Wang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303
| | - David Ostrov
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Nicolas Coant
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794
| | - Yusuf A Hannun
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794
| | - Aaron H Colby
- Ionic Pharmaceuticals, Brookline, MA 02445; and.,Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912; .,Georgia Cancer Center, Medical College of Georgia, Augusta, GA 30912.,Charlie Norwood VA Medical Center, Augusta, GA 30904
| |
Collapse
|
19
|
Zhang X, Sakamoto W, Canals D, Ishibashi M, Matsuda M, Nishida K, Toyoshima M, Shigeta S, Taniguchi M, Senkal CE, Okazaki T, Yaegashi N, Hannun YA, Nabe T, Kitatani K. Ceramide synthase 2-C 24:1 -ceramide axis limits the metastatic potential of ovarian cancer cells. FASEB J 2021; 35:e21287. [PMID: 33423335 PMCID: PMC8237407 DOI: 10.1096/fj.202001504rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/28/2020] [Accepted: 12/03/2020] [Indexed: 12/23/2022]
Abstract
Regulation of sphingolipid metabolism plays a role in cellular homeostasis, and dysregulation of these pathways is involved in cancer progression. Previously, our reports identified ceramide as an anti-metastatic lipid. In the present study, we investigated the biochemical alterations in ceramide-centered metabolism of sphingolipids that were associated with metastatic potential. We established metastasis-prone sublines of SKOV3 ovarian cancer cells using an in vivo selection method. These cells showed decreases in ceramide levels and ceramide synthase (CerS) 2 expression. Moreover, CerS2 downregulation in ovarian cancer cells promoted metastasis in vivo and potentiated cell motility and invasiveness. Moreover, CerS2 knock-in suppressed the formation of lamellipodia required for cell motility in this cell line. In order to define specific roles of ceramide species in cell motility controlled by CerS2, the effect of exogenous long- and very long-chain ceramide species on the formation of lamellipodia was evaluated. Treatment with distinct ceramides increased cellular ceramides and had inhibitory effects on the formation of lamellipodia. Interestingly, blocking the recycling pathway of ceramides by a CerS inhibitor was ineffective in the suppression of exogenous C24:1 -ceramide for the formation of lamellipodia. These results suggested that C24:1 -ceramide, a CerS2 metabolite, predominantly suppresses the formation of lamellipodia without the requirement for deacylation/reacylation. Moreover, knockdown of neutral ceramidase suppressed the formation of lamellipodia concomitant with upregulation of C24:1 -ceramide. Collectively, the CerS2-C24:1 -ceramide axis, which may be countered by neutral ceramidase, is suggested to limit cell motility and metastatic potential. These findings may provide insights that lead to further development of ceramide-based therapy and biomarkers for metastatic ovarian cancer.
Collapse
Affiliation(s)
- Xuewei Zhang
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Wataru Sakamoto
- Department of Medicine, Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Daniel Canals
- Department of Medicine, Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Masumi Ishibashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Masaya Matsuda
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Science, Setsunan University, Osaka, Japan
| | - Kentaro Nishida
- Department of Integrative Pharmaceutical Sciences, Faculty of Pharmaceutical Science, Setsunan University, Osaka, Japan
| | - Masafumi Toyoshima
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Shogo Shigeta
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Makoto Taniguchi
- Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Can E. Senkal
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, VA, USA
| | - Toshiro Okazaki
- Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
- Department of Hematology and Immunology, Kanazawa Medical University, Ishikawa, Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yusuf A. Hannun
- Department of Medicine, Stony Brook Cancer Center, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry, Stony Brook University, Stony Brook, NY, USA
| | - Takeshi Nabe
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Science, Setsunan University, Osaka, Japan
| | - Kazuyuki Kitatani
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Science, Setsunan University, Osaka, Japan
| |
Collapse
|
20
|
Caputo S, Di Martino S, Cilibrasi V, Tardia P, Mazzonna M, Russo D, Penna I, Summa M, Bertozzi SM, Realini N, Margaroli N, Migliore M, Ottonello G, Liu M, Lansbury P, Armirotti A, Bertorelli R, Ray SS, Skerlj R, Scarpelli R. Design, Synthesis, and Biological Evaluation of a Series of Oxazolone Carboxamides as a Novel Class of Acid Ceramidase Inhibitors. J Med Chem 2020; 63:15821-15851. [PMID: 33290061 PMCID: PMC7770833 DOI: 10.1021/acs.jmedchem.0c01561] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Acid
ceramidase (AC) is a cysteine hydrolase that plays a crucial
role in the metabolism of lysosomal ceramides, important members of
the sphingolipid family, a diversified class of bioactive molecules
that mediate many biological processes ranging from cell structural
integrity, signaling, and cell proliferation to cell death. In the
effort to expand the structural diversity of the existing collection
of AC inhibitors, a novel class of substituted oxazol-2-one-3-carboxamides
were designed and synthesized. Herein, we present the chemical optimization
of our initial hits, 2-oxo-4-phenyl-N-(4-phenylbutyl)oxazole-3-carboxamide 8a and 2-oxo-5-phenyl-N-(4-phenylbutyl)oxazole-3-carboxamide 12a, which resulted in the identification of 5-[4-fluoro-2-(1-methyl-4-piperidyl)phenyl]-2-oxo-N-pentyl-oxazole-3-carboxamide 32b as a potent
AC inhibitor with optimal physicochemical and metabolic properties,
showing target engagement in human neuroblastoma SH-SY5Y cells and
a desirable pharmacokinetic profile in mice, following intravenous
and oral administration. 32b enriches the arsenal of
promising lead compounds that may therefore act as useful pharmacological
tools for investigating the potential therapeutic effects of AC inhibition
in relevant sphingolipid-mediated disorders.
Collapse
Affiliation(s)
- Samantha Caputo
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Simona Di Martino
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Vincenzo Cilibrasi
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Piero Tardia
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Marco Mazzonna
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Debora Russo
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,D3-Pharma Chemistry, Via Morego 30, I-16163 Genova, Italy
| | - Ilaria Penna
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,D3-Pharma Chemistry, Via Morego 30, I-16163 Genova, Italy
| | - Maria Summa
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Analytical Chemistry and Translational Pharmacology, Via Morego 30, I-16163 Genova, Italy
| | - Sine Mandrup Bertozzi
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Analytical Chemistry and Translational Pharmacology, Via Morego 30, I-16163 Genova, Italy
| | - Natalia Realini
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Natasha Margaroli
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Marco Migliore
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Giuliana Ottonello
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Analytical Chemistry and Translational Pharmacology, Via Morego 30, I-16163 Genova, Italy
| | - Min Liu
- Lysosomal Therapeutics Inc., 19 Blackstone Street, Cambridge, Massachusetts 02139, United States
| | - Peter Lansbury
- Lysosomal Therapeutics Inc., 19 Blackstone Street, Cambridge, Massachusetts 02139, United States
| | - Andrea Armirotti
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Analytical Chemistry and Translational Pharmacology, Via Morego 30, I-16163 Genova, Italy
| | - Rosalia Bertorelli
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Analytical Chemistry and Translational Pharmacology, Via Morego 30, I-16163 Genova, Italy
| | - Soumya S Ray
- Lysosomal Therapeutics Inc., 19 Blackstone Street, Cambridge, Massachusetts 02139, United States
| | - Renato Skerlj
- Lysosomal Therapeutics Inc., 19 Blackstone Street, Cambridge, Massachusetts 02139, United States
| | - Rita Scarpelli
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| |
Collapse
|
21
|
Neutral ceramidase is a marker for cognitive performance in rats and monkeys. Pharmacol Rep 2020; 73:73-84. [PMID: 32936422 PMCID: PMC7862079 DOI: 10.1007/s43440-020-00159-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/20/2022]
Abstract
Background Ceramides are lipid molecules determining cell integrity and intercellular signaling, and thus, involved in the pathogenesis of several psychiatric and neurodegenerative disorders. However, little is known about the role of particular enzymes of the ceramide metabolism in the mechanisms of normal behavioral plasticity. Here, we studied the contribution of neutral ceramidase (NC), one of the main enzymes mediating ceramide degradation, in the mechanisms of learning and memory in rats and non-human primates. Methods Naïve Wistar rats and black tufted-ear marmosets (Callithrix penicillata) were tested in several tests for short- and long-term memory and then divided into groups with various memory performance. The activities of NC and acid ceramidase (AC) were measured in these animals. Additionally, anxiety and depression-like behavior and brain levels of monoamines were assessed in the rats. Results We observed a predictive role of NC activity in the blood serum for superior performance of long-term object memory tasks in both species. A brain area analysis suggested that high NC activity in the ventral mesencephalon (VM) predicts better short-term memory performance in rats. High NC activity in the VM was also associated with worse long-term object memory, which might be mediated by an enhanced depression-like state and a monoaminergic imbalance. Conclusions Altogether, these data suggest a role for NC in short- and long-term memory of various mammalian species. Serum activity of NC may possess a predictive role in the assessing the performance of certain types of memory. Electronic supplementary material The online version of this article (10.1007/s43440-020-00159-2) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Duarte C, Akkaoui J, Yamada C, Ho A, Mao C, Movila A. Elusive Roles of the Different Ceramidases in Human Health, Pathophysiology, and Tissue Regeneration. Cells 2020; 9:cells9061379. [PMID: 32498325 PMCID: PMC7349419 DOI: 10.3390/cells9061379] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 12/29/2022] Open
Abstract
Ceramide and sphingosine are important interconvertible sphingolipid metabolites which govern various signaling pathways related to different aspects of cell survival and senescence. The conversion of ceramide into sphingosine is mediated by ceramidases. Altogether, five human ceramidases—named acid ceramidase, neutral ceramidase, alkaline ceramidase 1, alkaline ceramidase 2, and alkaline ceramidase 3—have been identified as having maximal activities in acidic, neutral, and alkaline environments, respectively. All five ceramidases have received increased attention for their implications in various diseases, including cancer, Alzheimer’s disease, and Farber disease. Furthermore, the potential anti-inflammatory and anti-apoptotic effects of ceramidases in host cells exposed to pathogenic bacteria and viruses have also been demonstrated. While ceramidases have been a subject of study in recent decades, our knowledge of their pathophysiology remains limited. Thus, this review provides a critical evaluation and interpretive analysis of existing literature on the role of acid, neutral, and alkaline ceramidases in relation to human health and various diseases, including cancer, neurodegenerative diseases, and infectious diseases. In addition, the essential impact of ceramidases on tissue regeneration, as well as their usefulness in enzyme replacement therapy, is also discussed.
Collapse
Affiliation(s)
- Carolina Duarte
- Department of Periodontology, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33324, USA; (J.A.); (C.Y.); (A.H.)
- Correspondence: (C.D.); (A.M.); Tel.: +1-954-262-7306 (A.M.)
| | - Juliet Akkaoui
- Department of Periodontology, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33324, USA; (J.A.); (C.Y.); (A.H.)
| | - Chiaki Yamada
- Department of Periodontology, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33324, USA; (J.A.); (C.Y.); (A.H.)
| | - Anny Ho
- Department of Periodontology, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33324, USA; (J.A.); (C.Y.); (A.H.)
| | - Cungui Mao
- Department of Medicine, The State University of New York at Stony Brook, Stony Brook, NY 11794, USA;
- Cancer Center, The State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| | - Alexandru Movila
- Department of Periodontology, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33324, USA; (J.A.); (C.Y.); (A.H.)
- Institute for Neuro-Immune Medicine, Nova Southeastern University, Fort Lauderdale, FL 33324, USA
- Correspondence: (C.D.); (A.M.); Tel.: +1-954-262-7306 (A.M.)
| |
Collapse
|
23
|
Singh SK, Spiegel S. Sphingosine-1-phosphate signaling: A novel target for simultaneous adjuvant treatment of triple negative breast cancer and chemotherapy-induced neuropathic pain. Adv Biol Regul 2019; 75:100670. [PMID: 31708456 DOI: 10.1016/j.jbior.2019.100670] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 10/10/2019] [Accepted: 10/12/2019] [Indexed: 10/25/2022]
Abstract
Triple-negative breast cancer (TNBC) is very aggressive with high metastatic and mortality rates and unfortunately, except for chemotherapy, there are few therapeutic options. The bioactive sphingolipid metabolite sphingosine-1-phosphate (S1P) regulates numerous processes important for cancer progression, metastasis, and neuropathic pain. The pro-drug FTY720 (fingolimod, Gilenya) used to treat multiple sclerosis is phosphorylated in the body to a S1P mimic that binds to S1PRs, except S1PR2, and also acts as a functional antagonist of S1PR1. This review highlights current findings showing that FTY720 has multiple anti-cancer activities and simultaneously prevents formation and actions of S1P. Moreover, in mouse breast cancer models, treatment with FTY720 reduces tumor growth, metastasis, and enhances sensitivity of advanced and hormonal refractory breast cancer and TNBC to conventional therapies. We discuss recent studies demonstrating that neuropathic pain induced by the chemotherapeutic bortezomib is also greatly reduced by administration of clinically relevant doses of FTY720, likely by targeting S1PR1 on astrocytes. FTY720 also shows promising anticancer potential in pre-clinical studies and is FDA approved, thus we suggest in this review that further studies are needed to pave the way for fast-tracking approval of FTY720/fingolimod for enhancing chemotherapy effectiveness and reduction of painful neuropathies.
Collapse
Affiliation(s)
- Sandeep K Singh
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine and the Massey Cancer Center, Richmond, VA, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine and the Massey Cancer Center, Richmond, VA, USA.
| |
Collapse
|
24
|
Coant N, Hannun YA. Neutral ceramidase: Advances in mechanisms, cell regulation, and roles in cancer. Adv Biol Regul 2018; 71:141-146. [PMID: 30389354 DOI: 10.1016/j.jbior.2018.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 01/11/2023]
Abstract
Extensive research conducted in the last three decades has identified the roles for the main bioactive sphingolipids, namely ceramide, sphingosine, and sphingosine 1-phosphate (S1P) as key regulators of cellular homeostasis, growth and death. One of the major groups of enzymes in the ceramide pathway, ceramidases, converts ceramide into sphingosine and fatty acids, with sphingosine being further metabolized to S1P. Thus, these enzymes play important roles in the network controlling the functions associated with these bioactive sphingolipids. Among the family of ceramidases, neutral ceramidase (nCDase), which is named according to its optimal pH for catalytic activity, has received increased attention in the last decade. The goal of this review is to provide a brief background on bioactive sphingolipids and the ceramidases. We then describe more recent advances on nCDase, specifically the resolution of its crystal structure and understanding its roles in cell biology and physiology.
Collapse
Affiliation(s)
- Nicolas Coant
- Health Science Center, Stony Brook University, 100 Nicolls Road, T15, 023, 11794, Stony Brook, NY, USA.
| | - Yusuf A Hannun
- Health Science Center, Stony Brook University, 100 Nicolls Road, L4, 182, 11794, Stony Brook, NY, USA.
| |
Collapse
|