1
|
Chin T, Lee XE, Ng PY, Lee Y, Dreesen O. The role of cellular senescence in skin aging and age-related skin pathologies. Front Physiol 2023; 14:1297637. [PMID: 38074322 PMCID: PMC10703490 DOI: 10.3389/fphys.2023.1297637] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/07/2023] [Indexed: 12/21/2024] Open
Abstract
Aging is the result of a gradual functional decline at the cellular, and ultimately, organismal level, resulting in an increased risk of developing a variety of chronic illnesses, such as cardiovascular disease, stroke, cancer and diabetes. The skin is the largest organ of the human body, and the site where signs of aging are most visible. These signs include thin and dry skin, sagging, loss of elasticity, wrinkles, as well as aberrant pigmentation. The appearance of these features is accelerated by exposure to extrinsic factors such as ultraviolet (UV) radiation or pollution, as well as intrinsic factors including time, genetics, and hormonal changes. At the cellular level, aging is associated with impaired proteostasis and an accumulation of macromolecular damage, genomic instability, chromatin reorganization, telomere shortening, remodelling of the nuclear lamina, proliferation defects and premature senescence. Cellular senescence is a state of permanent growth arrest and a key hallmark of aging in many tissues. Due to their inability to proliferate, senescent cells no longer contribute to tissue repair or regeneration. Moreover, senescent cells impair tissue homeostasis, promote inflammation and extracellular matrix (ECM) degradation by secreting molecules collectively known as the "senescence-associated secretory phenotype" (SASP). Senescence can be triggered by a number of different stimuli such as telomere shortening, oncogene expression, or persistent activation of DNA damage checkpoints. As a result, these cells accumulate in aging tissues, including human skin. In this review, we focus on the role of cellular senescence during skin aging and the development of age-related skin pathologies, and discuss potential strategies to rejuvenate aged skin.
Collapse
Affiliation(s)
- Toby Chin
- Lee Kong Chiang School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Xin Er Lee
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Pei Yi Ng
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yaelim Lee
- Mechanobiology Institute, National University of Singapore, T-Lab, Singapore, Singapore
| | - Oliver Dreesen
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, T-Lab, Singapore, Singapore
| |
Collapse
|
2
|
Velazquez FN, Stith JL, Zhang L, Allam AM, Haley J, Obeid LM, Snider AJ, Hannun YA. Targeting sphingosine kinase 1 in p53KO thymic lymphoma. FASEB J 2023; 37:e23247. [PMID: 37800872 PMCID: PMC11740227 DOI: 10.1096/fj.202301417r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023]
Abstract
Sphingosine kinase 1 (SK1) is a key sphingolipid enzyme that is upregulated in several types of cancer, including lymphoma which is a heterogenous group of malignancies. Treatment for lymphoma has improved significantly by the introduction of new therapies; however, subtypes with tumor protein P53 (p53) mutations or deletion have poor prognosis, making it critical to explore new therapeutic strategies in this context. SK1 has been proposed as a therapeutic target in different types of cancer; however, the effect of targeting SK1 in cancers with p53 deletion has not been evaluated yet. Previous work from our group suggests that loss of SK1 is a key event in mediating the tumor suppressive effect of p53. Employing both genetic and pharmacological approaches to inhibit SK1 function in Trp53KO mice, we show that targeting SK1 decreases tumor growth of established p53KO thymic lymphoma. Inducible deletion of Sphk1 or its pharmacological inhibition drive increased cell death in tumors which is accompanied by selective accumulation of sphingosine levels. These results demonstrate the relevance of SK1 in the growth and maintenance of lymphoma in the absence of p53 function, positioning this enzyme as a potential therapeutic target for the treatment of tumors that lack functional p53.
Collapse
Affiliation(s)
- Fabiola N Velazquez
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - Jeffrey L Stith
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - Leiqing Zhang
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - Amira M Allam
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - John Haley
- Biological Mass Spectrometry Center, Stony Brook Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Lina M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - Ashley J Snider
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, and University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
3
|
Djokovic N, Djuric A, Ruzic D, Srdic-Rajic T, Nikolic K. Correlating Basal Gene Expression across Chemical Sensitivity Data to Screen for Novel Synergistic Interactors of HDAC Inhibitors in Pancreatic Carcinoma. Pharmaceuticals (Basel) 2023; 16:294. [PMID: 37259439 PMCID: PMC9964546 DOI: 10.3390/ph16020294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/23/2023] [Accepted: 02/11/2023] [Indexed: 11/25/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal malignancies. Development of the chemoresistance in the PDAC is one of the key contributors to the poor survival outcomes and the major reason for urgent development of novel pharmacological approaches in a treatment of PDAC. Systematically tailored combination therapy holds the promise for advancing the treatment of PDAC. However, the number of possible combinations of pharmacological agents is too large to be explored experimentally. In respect to the many epigenetic alterations in PDAC, epigenetic drugs including histone deacetylase inhibitors (HDACi) could be seen as the game changers especially in combined therapy settings. In this work, we explored a possibility of using drug-sensitivity data together with the basal gene expression of pancreatic cell lines to predict combinatorial options available for HDACi. Developed bioinformatics screening protocol for predictions of synergistic drug combinations in PDAC identified the sphingolipid signaling pathway with associated downstream effectors as a promising novel targets for future development of multi-target therapeutics or combined therapy with HDACi. Through the experimental validation, we have characterized novel synergism between HDACi and a Rho-associated protein kinase (ROCK) inhibitor RKI-1447, and between HDACi and a sphingosine 1-phosphate (S1P) receptor agonist fingolimod.
Collapse
Affiliation(s)
- Nemanja Djokovic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Ana Djuric
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Tatjana Srdic-Rajic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia
| |
Collapse
|
4
|
Hamsanathan S, Gurkar AU. Lipids as Regulators of Cellular Senescence. Front Physiol 2022; 13:796850. [PMID: 35370799 PMCID: PMC8965560 DOI: 10.3389/fphys.2022.796850] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
Abstract
Lipids are key macromolecules that perform a multitude of biological functions ranging from maintaining structural integrity of membranes, energy storage, to signaling molecules. Unsurprisingly, variations in lipid composition and its levels can influence the functional and physiological state of the cell and its milieu. Cellular senescence is a permanent state of cell cycle arrest and is a hallmark of the aging process, as well as several age-related pathologies. Senescent cells are often characterized by alterations in morphology, metabolism, chromatin remodeling and exhibit a complex pro-inflammatory secretome (SASP). Recent studies have shown that the regulation of specific lipid species play a critical role in senescence. Indeed, some lipid species even contribute to the low-grade inflammation associated with SASP. Many protein regulators of senescence have been well characterized and are associated with lipid metabolism. However, the link between critical regulators of cellular senescence and senescence-associated lipid changes is yet to be elucidated. Here we systematically review the current knowledge on lipid metabolism and dynamics of cellular lipid content during senescence. We focus on the roles of major players of senescence in regulating lipid metabolism. Finally, we explore the future prospects of lipid research in senescence and its potential to be targeted as senotherapeutics.
Collapse
Affiliation(s)
- Shruthi Hamsanathan
- Aging Institute of UPMC, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Aditi U. Gurkar
- Aging Institute of UPMC, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Geriatric Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, United States
- *Correspondence: Aditi U. Gurkar,
| |
Collapse
|
5
|
Elhady SS, Habib ES, Abdelhameed RFA, Goda MS, Hazem RM, Mehanna ET, Helal MA, Hosny KM, Diri RM, Hassanean HA, Ibrahim AK, Eltamany EE, Abdelmohsen UR, Ahmed SA. Anticancer Effects of New Ceramides Isolated from the Red Sea Red Algae Hypnea musciformis in a Model of Ehrlich Ascites Carcinoma: LC-HRMS Analysis Profile and Molecular Modeling. Mar Drugs 2022; 20:md20010063. [PMID: 35049918 PMCID: PMC8778197 DOI: 10.3390/md20010063] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 02/04/2023] Open
Abstract
Different classes of phytochemicals were previously isolated from the Red Sea algae Hypnea musciformis as sterols, ketosteroids, fatty acids, and terpenoids. Herein, we report the isolation of three fatty acids-docosanoic acid 4, hexadecenoic acid 5, and alpha hydroxy octadecanoic acid 6-as well as three ceramides-A (1), B (2), and C (3)-with 9-methyl-sphinga-4,8-dienes and phytosphingosine bases. Additionally, different phytochemicals were determined using the liquid chromatography coupled with electrospray ionization high-resolution mass spectrometry (LC-ESI-HRMS) technique. Ceramides A (1) and B (2) exhibited promising in vitro cytotoxic activity against the human breast adenocarcinoma (MCF-7) cell line when compared with doxorubicin as a positive control. Further in vivo study and biochemical estimation in a mouse model of Ehrlich ascites carcinoma (EAC) revealed that both ceramides A (1) and B (2) at doses of 1 and 2 mg/kg, respectively, significantly decreased the tumor size in mice inoculated with EAC cells. The higher dose (2 mg/kg) of ceramide B (2) particularly expressed the most pronounced decrease in serum levels of vascular endothelial growth factor -B (VEGF-B) and tumor necrosis factor-α (TNF-α) markers, as well as the expression levels of the growth factor midkine in tumor tissue relative to the EAC control group. The highest expression of apoptotic factors, p53, Bax, and caspase 3 was observed in the same group that received 2 mg/kg of ceramide B (2). Molecular docking simulations suggested that ceramides A (1) and B (2) could bind in the deep grove between the H2 helix and the Ser240-P250 loop of p53, preventing its interaction with MDM2 and leading to its accumulation. In conclusion, this study reports the cytotoxic, apoptotic, and antiangiogenic effects of ceramides isolated from the Red Sea algae Hypnea musciformis in an experimental model of EAC.
Collapse
Affiliation(s)
- Sameh S. Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Eman S. Habib
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.S.H.); (M.S.G.); (H.A.H.); (A.K.I.); (E.E.E.)
| | - Reda F. A. Abdelhameed
- Department of Pharmacognosy, Faculty of Pharmacy, Galala University, New Galala 43713, Egypt;
| | - Marwa S. Goda
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.S.H.); (M.S.G.); (H.A.H.); (A.K.I.); (E.E.E.)
| | - Reem M. Hazem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| | - Eman T. Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt;
| | - Mohamed A. Helal
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, October Gardens, 6th of October, Giza 12578, Egypt;
- Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Khaled M. Hosny
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Reem M. Diri
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Hashim A. Hassanean
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.S.H.); (M.S.G.); (H.A.H.); (A.K.I.); (E.E.E.)
| | - Amany K. Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.S.H.); (M.S.G.); (H.A.H.); (A.K.I.); (E.E.E.)
| | - Enas E. Eltamany
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.S.H.); (M.S.G.); (H.A.H.); (A.K.I.); (E.E.E.)
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt;
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, New Minia 61111, Egypt
| | - Safwat A. Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (E.S.H.); (M.S.G.); (H.A.H.); (A.K.I.); (E.E.E.)
- Correspondence: or ; Tel.: +20-010-92638387
| |
Collapse
|
6
|
Chen LL, Wang WJ. p53 regulates lipid metabolism in cancer. Int J Biol Macromol 2021. [DOI: https://doi.org/10.1016/j.ijbiomac.2021.09.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
7
|
Chen LL, Wang WJ. p53 regulates lipid metabolism in cancer. Int J Biol Macromol 2021; 192:45-54. [PMID: 34619274 DOI: 10.1016/j.ijbiomac.2021.09.188] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/22/2021] [Accepted: 09/28/2021] [Indexed: 02/07/2023]
Abstract
Reprogrammed cell metabolism is a well-accepted hallmark of cancer. Metabolism changes provide energy and precursors for macromolecule biosynthesis to satisfy the survival needs of cancer cells. The specific changes in different aspects of lipid metabolism in cancer cells have been focused in recent years. These changes can affect cell growth, proliferation, differentiation and motility through affecting membranes synthesis, energy homeostasis and cell signaling. The tumor suppressor p53 plays vital roles in the control of cell proliferation, senescence, DNA repair, and cell death in cancer through various transcriptional and non-transcriptional activities. Accumulating evidences indicate that p53 also regulates cellular metabolism, which appears to contribute to its tumor suppressive functions. Particularly the role of p53 in regulating lipid metabolism has gained more and more attention in recent decades. In this review, we summarize recent advances in the function of p53 on lipid metabolism in cancer. Further understanding and research on the role of p53 in lipid metabolism regulation will provide a potential therapeutic window for cancer treatment.
Collapse
Affiliation(s)
- Ling-Li Chen
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Wen-Jun Wang
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China.
| |
Collapse
|
8
|
Barron KA, Jeffries KA, Krupenko NI. Sphingolipids and the link between alcohol and cancer. Chem Biol Interact 2020; 322:109058. [PMID: 32171848 DOI: 10.1016/j.cbi.2020.109058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/20/2019] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Epidemiological evidence underscores alcohol consumption as a strong risk factor for multiple cancer types, with liver cancer being most commonly associated with alcohol intake. While mechanisms linking alcohol consumption to malignant tumor development are not fully understood, the likely players in ethanol-induced carcinogenesis are genotoxic stress caused by formation of acetaldehyde, increased oxidative stress, and altered nutrient metabolism, including the impairment of methyl transfer reactions. Alterations of sphingolipid metabolism and associated signaling pathways are another potential link between ethanol and cancer development. In particular, ceramides are involved in the regulation of cellular proliferation, differentiation, senescence, and apoptosis and are known to function as important regulators of malignant transformation as well as tumor progression. However, to date, the cross-talk between ceramides and alcohol in cancer disease is largely an open question and only limited data are available on this subject. Most studies linking ceramide to cancer considered liver steatosis as the underlying mechanism, which is not surprising taking into consideration that ceramide pathways are an integral part of the overall lipid metabolism. This review summarizes the latest studies pointing to ceramide as an important mediator of cancer-promoting effects of chronic alcohol consumption and underscores the necessity of understanding the role of sphingolipids and lipid signaling in response to alcohol in order to prevent and/or successfully manage diseases caused by alcohol.
Collapse
Affiliation(s)
| | | | - Natalia I Krupenko
- Department of Nutrition, UNC Chapel Hill, USA; Nutrition Research Institute, UNC Chapel Hill, USA.
| |
Collapse
|
9
|
Brachtendorf S, El-Hindi K, Grösch S. WITHDRAWN: Ceramide synthases in cancer therapy and chemoresistance. Prog Lipid Res 2019:100992. [PMID: 31442523 DOI: 10.1016/j.plipres.2019.100992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 10/26/2022]
Affiliation(s)
- Sebastian Brachtendorf
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern Kai 7, Frankfurt 60590, Germany
| | - Khadija El-Hindi
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern Kai 7, Frankfurt 60590, Germany
| | - Sabine Grösch
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern Kai 7, Frankfurt 60590, Germany
| |
Collapse
|
10
|
Brachtendorf S, El-Hindi K, Grösch S. Ceramide synthases in cancer therapy and chemoresistance. Prog Lipid Res 2019; 74:160-185. [DOI: 10.1016/j.plipres.2019.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/24/2022]
|
11
|
C 16-ceramide is a natural regulatory ligand of p53 in cellular stress response. Nat Commun 2018; 9:4149. [PMID: 30297838 PMCID: PMC6175828 DOI: 10.1038/s41467-018-06650-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 09/12/2018] [Indexed: 12/22/2022] Open
Abstract
Ceramides are important participants of signal transduction, regulating fundamental cellular processes. Here we report the mechanism for activation of p53 tumor suppressor by C16-ceramide. C16-ceramide tightly binds within the p53 DNA-binding domain (Kd ~ 60 nM), in close vicinity to the Box V motif. This interaction is highly selective toward the ceramide acyl chain length with its C10 atom being proximal to Ser240 and Ser241. Ceramide binding stabilizes p53 and disrupts its complex with E3 ligase MDM2 leading to the p53 accumulation, nuclear translocation and activation of the downstream targets. This mechanism of p53 activation is fundamentally different from the canonical p53 regulation through protein–protein interactions or posttranslational modifications. The discovered mechanism is triggered by serum or folate deprivation implicating it in the cellular response to nutrient/metabolic stress. Our study establishes C16-ceramide as a natural small molecule activating p53 through the direct binding. Ceramides are important participants of signal transduction, regulating fundamental cellular processes. Here authors show that C16-ceramide binds to the tumor suppressor p53, disrupts its interaction with MDM2 and facilitates p53 accumulation and activation of its downstream targets.
Collapse
|
12
|
Brachtendorf S, Wanger RA, Birod K, Thomas D, Trautmann S, Wegner MS, Fuhrmann DC, Brüne B, Geisslinger G, Grösch S. Chemosensitivity of human colon cancer cells is influenced by a p53-dependent enhancement of ceramide synthase 5 and induction of autophagy. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1214-1227. [PMID: 30059758 DOI: 10.1016/j.bbalip.2018.07.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/03/2018] [Accepted: 07/24/2018] [Indexed: 12/21/2022]
Abstract
Resistance against chemotherapy is a life-threatening complication in colon cancer therapy. To increase response rate, new additional targets that contribute to chemoresistance are still needed to be explored. Ceramides, which belong to the group of sphingolipids, are well-known regulators of cell death and survival, respectively. Here, we show that in human wild-type (wt) p53 HCT-116 colon cancer cells treatment with oxaliplatin or 5-fluorouracil (5-FU) leads to a strong increase in ceramide synthase 5 (CerS5) expression and C16:0-ceramide levels, which was not shown in HCT-116 lacking p53 expression (HCT-116 p53-/-). The increase in CerS5 expression occurs by stabilizing CerS5 mRNA at the 3'-UTR. By contrast, in the p53-deficient cells CerS2 expression and CerS2-related C24:0- and C24:1-ceramide levels were elevated which is possibly related to enhanced polyadenylation of the CerS2 transcript in these cells. Stable knockdown of CerS5 expression using CerS5-targeting shRNA led to an increased sensitivity of HCT-116 p53wt cells, but not of p53-/- cells, to oxaliplatin and 5-FU. Enhanced sensitivity was accompanied by an inhibition of autophagy and inhibition of mitochondrial respiration in these cells. However, knockdown of CerS2 had no significant effects on chemosensitivity of both cell lines. In conclusion, in p53wt colon cancer cells chemosensitivity against oxaliplatin or 5-FU could be enhanced by downregulation of CerS5 expression leading to reduced autophagy and mitochondrial respiration.
Collapse
Affiliation(s)
- Sebastian Brachtendorf
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Germany
| | - Ruth Anna Wanger
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Germany
| | - Kerstin Birod
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Germany
| | - Sandra Trautmann
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Germany
| | - Marthe-Susanna Wegner
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Germany
| | - Dominik C Fuhrmann
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine and Pharmacology (TMP), Frankfurt, Germany
| | - Sabine Grösch
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Germany.
| |
Collapse
|
13
|
Onyango AN. Cellular Stresses and Stress Responses in the Pathogenesis of Insulin Resistance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4321714. [PMID: 30116482 PMCID: PMC6079365 DOI: 10.1155/2018/4321714] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Accepted: 02/18/2018] [Indexed: 12/14/2022]
Abstract
Insulin resistance (IR), a key component of the metabolic syndrome, precedes the development of diabetes, cardiovascular disease, and Alzheimer's disease. Its etiological pathways are not well defined, although many contributory mechanisms have been established. This article summarizes such mechanisms into the hypothesis that factors like nutrient overload, physical inactivity, hypoxia, psychological stress, and environmental pollutants induce a network of cellular stresses, stress responses, and stress response dysregulations that jointly inhibit insulin signaling in insulin target cells including endothelial cells, hepatocytes, myocytes, hypothalamic neurons, and adipocytes. The insulin resistance-inducing cellular stresses include oxidative, nitrosative, carbonyl/electrophilic, genotoxic, and endoplasmic reticulum stresses; the stress responses include the ubiquitin-proteasome pathway, the DNA damage response, the unfolded protein response, apoptosis, inflammasome activation, and pyroptosis, while the dysregulated responses include the heat shock response, autophagy, and nuclear factor erythroid-2-related factor 2 signaling. Insulin target cells also produce metabolites that exacerbate cellular stress generation both locally and systemically, partly through recruitment and activation of myeloid cells which sustain a state of chronic inflammation. Thus, insulin resistance may be prevented or attenuated by multiple approaches targeting the different cellular stresses and stress responses.
Collapse
Affiliation(s)
- Arnold N. Onyango
- Department of Food Science and Technology, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, Nairobi 00200, Kenya
| |
Collapse
|
14
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a group of liver disorders encompassing simple hepatic steatosis and its more aggressive forms of nonalcoholic steatohepatitis and cirrhosis. It is a rapidly growing health concern and the major cause for the increasing incidence of primary liver tumors. Unequivocal evidence shows that sphingolipid metabolism is altered in the course of the disease and these changes might contribute to NAFLD progression. Recent data provide solid support to the notion that deregulated ceramide and sphingosine-1-phosphate metabolism are present at all stages of NAFLD, i.e., steatosis, nonalcoholic steatohepatitis, advanced fibrosis, and hepatocellular carcinoma (HCC). Insulin sensitivity, de novo lipogenesis, and the resulting lipotoxicity, fibrosis, and angiogenesis are all seemingly regulated in a manner that involves either ceramide and/or sphingosine-1-phosphate. Sphingolipids might also participate in the onset of hepatocellular senescence. The latter has been shown to contribute to the advancement of cirrhosis to HCC in the classical cases of end-stage liver disease, i.e., viral- or alcohol-induced; however, emerging evidence suggests that senescence is also involved in the pathogenicity of NAFLD possibly via changes in ceramide metabolism.
Collapse
|
15
|
Abstract
Ceramides, important players in signal transduction, interact with multiple cellular pathways, including p53 pathways. However, the relationship between ceramide and p53 is very complex, and mechanisms underlying their coregulation are diverse and not fully characterized. The role of p53, an important cellular regulator and a transcription factor, is linked to its tumor suppressor function. Ceramides are involved in the regulation of fundamental processes in cancer cells including cell death, proliferation, autophagy, and drug resistance. This regulation, however, can be pro-death or pro-survival depending on cancer type, the balance between ceramide species, the rate of their synthesis and utilization, and the availability of a specific array of downstream targets. This chapter highlights the central role of ceramide in sphingolipid metabolism, its role in cancer, specific effectors in ceramide pathways controlled by p53, and coregulation of ceramide and p53 signaling. We discuss the recent studies, which underscore the function of p53 in the regulation of ceramide pathways and the reciprocal regulation of p53 by ceramide. This complex relationship is based on several molecular mechanisms including the p53-dependent transcriptional regulation of enzymes in sphingolipid pathways, the activation of mutant p53 through ceramide-mediated alternative splicing, as well as modulation of the p53 function through direct and indirect effects on p53 coregulators and downstream targets. Further insight into the connections between ceramide and p53 will allow simultaneous targeting of the two pathways with a potential to yield more efficient anticancer therapeutics.
Collapse
Affiliation(s)
- Kristen A Jeffries
- Nutrition Research Institute, UNC Chapel Hill, Kannapolis, NC, United States
| | - Natalia I Krupenko
- Nutrition Research Institute, UNC Chapel Hill, Kannapolis, NC, United States; Department of Nutrition, UNC Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
16
|
Sun J, Jing S, Jiang R, Wang C, Zhang C, Chen J, Li H. Metabolomics study of the therapeutic mechanism of Schisandra chinensis lignans on aging rats induced by d-galactose. Clin Interv Aging 2018; 13:829-841. [PMID: 29750025 PMCID: PMC5935080 DOI: 10.2147/cia.s163275] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective The aim of this study was to evaluate the antiaging effect of Schisandra chinensis lignans (SCL) by analyzing the characteristics in the serum of d-galactose (d-gal)-induced rats. Methods Forty male Wistar rats were randomly divided into control group, d-gal model group, low-dose SCL group (50 mg/kg/d), medium-dose SCL group (100 mg/kg/d), and high-dose SCL group (200 mg/kg/d). A serum metabolomics analysis method based on rapid resolution liquid chromatography coupled with quadruple-time-of-flight mass spectrometry was carried out to study the characteristics of d-gal-induced aging rats and evaluate the antiaging effects of SCL, and multivariate statistical analysis was performed for pattern recognition and characteristic metabolites identification. The relative levels of p19, p53, and p21 genes in the brain tissue were measured by quantitative real-time polymerase chain reaction for investigating the underlying mechanism. Results Metabolomics analysis showed that 15 biomarkers were identified and 13 of them recovered to the normal levels after the administration of SCL. Based on the pathway analysis, the antiaging mechanisms of SCL might be involved in the following metabolic pathways: energy, amino acid, lipid, and phospholipid metabolism. Furthermore, SCL significantly inhibited the mRNA expression level of p19, p53, and p21 in the brain of aging rats induced by d-gal. Conclusion These results suggest that SCL can delay rat aging induced by d-gal through multiple pathways.
Collapse
Affiliation(s)
- Jinghui Sun
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, People's Republic of China
| | - Shu Jing
- General Surgery Area, Affiliated Hospital of Beihua University, Jilin, People's Republic of China
| | - Rui Jiang
- Jilin Technology Innovation Center for Chinese Medicine Biotechnology, College of Chemistry and Biology, Beihua University, Jilin, People's Republic of China
| | - Chunmei Wang
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, People's Republic of China
| | - Chengyi Zhang
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, People's Republic of China
| | - Jianguang Chen
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, People's Republic of China
| | - He Li
- Department of Pharmacology, College of Pharmacy, Beihua University, Jilin, People's Republic of China
| |
Collapse
|
17
|
Wang Y, Zhang C, Jin Y, Wang, He Q, Liu Z, Ai Q, Lei Y, Li Y, Song F, Bu Y. Alkaline ceramidase 2 is a novel direct target of p53 and induces autophagy and apoptosis through ROS generation. Sci Rep 2017; 7:44573. [PMID: 28294157 PMCID: PMC5353723 DOI: 10.1038/srep44573] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/09/2017] [Indexed: 12/22/2022] Open
Abstract
ACER2 is a critical sphingolipid metabolizing enzyme, and has been shown to be remarkably up-regulated following various stimuli such as DNA damage. However, the transcriptional regulatory mechanism of ACER2 gene and its potential role in the regulation of autophagy remain unknown. In this study, we have for the first time identified the human ACER2 gene promoter, and found that human ACER2 transcription is directly regulated by p53 and ACER2 is implicated in the induction of autophagy as well as apoptosis. A series of luciferase reporter assay demonstrated that ACER2 major promoter is located within its first intron where the consensus p53-binding sites exist. Consistently, forced expression of p53 significantly stimulated ACER2 transcription. Notably, p53-mediated autophagy and apoptosis were markedly enhanced by ACER2. Depletion of the essential autophagy gene ATG5 revealed that ACER2-induced autophagy facilitates its effect on apoptosis. Further studies clearly showed that ACER2-mediated autophagy and apoptosis are accompanied by ROS generation. In summary, our present study strongly suggests that ACER2 plays a pivotal role in p53-induced autophagy and apoptosis, and thus might serve as a novel and attractive molecular target for cancer treatment.
Collapse
Affiliation(s)
- Yitao Wang
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Chunxue Zhang
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Yuelei Jin
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Wang
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Qing He
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhu Liu
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Qing Ai
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Yi Li
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Fangzhou Song
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Youquan Bu
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
18
|
Denamur S, Boland L, Beyaert M, Verstraeten SL, Fillet M, Tulkens PM, Bontemps F, Mingeot-Leclercq MP. Subcellular mechanisms involved in apoptosis induced by aminoglycoside antibiotics: Insights on p53, proteasome and endoplasmic reticulum. Toxicol Appl Pharmacol 2016; 309:24-36. [DOI: 10.1016/j.taap.2016.08.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/20/2016] [Accepted: 08/22/2016] [Indexed: 12/21/2022]
|
19
|
Fekry B, Jeffries KA, Esmaeilniakooshkghazi A, Ogretmen B, Krupenko SA, Krupenko NI. CerS6 Is a Novel Transcriptional Target of p53 Protein Activated by Non-genotoxic Stress. J Biol Chem 2016; 291:16586-96. [PMID: 27302066 PMCID: PMC4974374 DOI: 10.1074/jbc.m116.716902] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Indexed: 12/19/2022] Open
Abstract
Our previous study suggested that ceramide synthase 6 (CerS6), an enzyme in sphingolipid biosynthesis, is regulated by p53: CerS6 was elevated in several cell lines in response to transient expression of p53 or in response to folate stress, which is known to activate p53. It was not clear, however, whether CerS6 gene is a direct transcriptional target of p53 or whether this was an indirect effect through additional regulatory factors. In the present study, we have shown that the CerS6 promoter is activated by p53 in luciferase assays, whereas transcriptionally inactive R175H p53 mutant failed to induce the luciferase expression from this promoter. In vitro immunoprecipitation assays and gel shift analyses have further demonstrated that purified p53 binds within the CerS6 promoter sequence spanning 91 bp upstream and 60 bp downstream of the transcription start site. The Promo 3.0.2 online tool for the prediction of transcription factor binding sites indicated the presence of numerous putative non-canonical p53 binding motifs in the CerS6 promoter. Luciferase assays and gel shift analysis have identified a single motif upstream of the transcription start as a key p53 response element. Treatment of cells with Nutlin-3 or low concentrations of actinomycin D resulted in a strong elevation of CerS6 mRNA and protein, thus demonstrating that CerS6 is a component of the non-genotoxic p53-dependent cellular stress response. This study has shown that by direct transcriptional activation of CerS6, p53 can regulate specific ceramide biosynthesis, which contributes to the pro-apoptotic cellular response.
Collapse
Affiliation(s)
- Baharan Fekry
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081
| | - Kristen A Jeffries
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081
| | - Amin Esmaeilniakooshkghazi
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081
| | - Besim Ogretmen
- the Department of Biochemistry and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425, and
| | - Sergey A Krupenko
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081, the Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Natalia I Krupenko
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081, the Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
20
|
Go H, Kim PJ, Jeon YK, Cho YM, Kim K, Park BH, Ku JY. Sphingosine-1-phosphate receptor 1 (S1PR1) expression in non-muscle invasive urothelial carcinoma: Association with poor clinical outcome and potential therapeutic target. Eur J Cancer 2015; 51:1937-45. [PMID: 26238015 DOI: 10.1016/j.ejca.2015.07.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/15/2015] [Accepted: 07/16/2015] [Indexed: 12/30/2022]
Abstract
AIM Sphingosine-1-phosphate receptor 1 (S1PR1) promotes tumour cell survival, invasion, anti-apoptosis, metastasis and radio/chemo-resistance in various cancers. However, the expression pattern and prognostic implications of S1PR1 in urothelial carcinoma remain unclear and thus were addressed here. METHODS Tissue microarrays composed of 395 initially diagnosed and transurethral resected urothelial carcinomas of the urinary bladder were immunostained for S1PR1 and phosphor-signal transducer and activator of transcription 3 (pSTAT3). S1PR1 expression was analysed according to clinicopathological features, expression of several anti-apoptosis/proliferation-related markers and patient's survival. RESULTS S1PR1 positivity was observed in 45.3% of urothelial carcinomas. Among patients with non-muscle invasive urothelial carcinoma (NMIC), S1PR1 positivity was associated with higher grade (P<0.001), higher subepithelial invasive component (P=0.006), lower papillary component (P=0.002), presence of metastasis (P=0.042) and high cancer-specific death (P<0.001). S1PR1 expression was correlated with pSTAT3 (P<0.001), survivin (P=0.008) and Ki-67 (P<0.001) expression. S1PR1 positivity predicted a shorter cancer-specific survival (CSS) in NMICs (P<0.001) and stage T1/high grade (T1HG) tumours (P=0.002). The Cox multivariate model was composed of S1PR1, survivin, lymphovascular invasion and age, and C-index was 0.781. S1PR1 positivity was correlated with shorter CSS in p53-positive T1HG carcinoma (P=0.003) in contrast to p53-negative T1HG carcinoma (P=0.205). In p53-overexpressing NMIC, S1PR1 was the only variable of the survival model and the C-index was 0.719. CONCLUSIONS S1PR1 expression was associated with unfavourable clinicopathological features and the expression of several anti-apoptosis/proliferation-related markers in urothelial carcinoma. S1PR1 serves as an independent predictor of cancer-specific death in NMIC. The model including S1PR1 showed highly accurate prediction for CSS in NMIC patients regardless of the modality of adjuvant therapy.
Collapse
Affiliation(s)
- Heounjeong Go
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea.
| | - Pil-Jong Kim
- Biomedical Knowledge Engineering Laboratory, Seoul National University, School of Dentistry, Seoul 110-749, Republic of Korea.
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Republic of Korea.
| | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea.
| | - Kyungeun Kim
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 110-746, Republic of Korea.
| | - Bong-Hee Park
- Department of Pathology, Green Cross Laboratories, Yongin-si, Kyunggi-do 446-770, Republic of Korea.
| | - Ja Yoon Ku
- University of Ulsan College of Medicine, Seoul 138-736, Republic of Korea.
| |
Collapse
|
21
|
Beyoğlu D, Krausz KW, Martin J, Maurhofer O, Dorow J, Ceglarek U, Gonzalez FJ, Dufour JF, Idle JR. Disruption of tumor suppressor gene Hint1 leads to remodeling of the lipid metabolic phenotype of mouse liver. J Lipid Res 2014; 55:2309-19. [PMID: 25193995 DOI: 10.1194/jlr.m050682] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A lipidomic and metabolomic investigation of serum and liver from mice was performed to gain insight into the tumor suppressor gene Hint1. A major reprogramming of lipid homeostasis was found in both serum and liver of Hint1-null (Hint(-/-)) mice, with significant changes in the levels of many lipid molecules, as compared with gender-, age-, and strain-matched WT mice. In the Hint1(-/-) mice, serum total and esterified cholesterol were reduced 2.5-fold, and lysophosphatidylcholines (LPCs) and lysophosphatidic acids were 10-fold elevated in serum, with a corresponding fall in phosphatidylcholines (PCs). In the liver, MUFAs and PUFAs, including arachidonic acid (AA) and its metabolic precursors, were also raised, as was mRNA encoding enzymes involved in AA de novo synthesis. There was also a significant 50% increase in hepatic macrophages in the Hint1(-/-) mice. Several hepatic ceramides and acylcarnitines were decreased in the livers of Hint1(-/-) mice. The changes in serum LPCs and PCs were neither related to hepatic phospholipase A2 activity nor to mRNAs encoding lysophosphatidylcholine acetyltransferases 1-4. The lipidomic phenotype of the Hint1(-/-) mouse revealed decreased inflammatory eicosanoids with elevated proliferative mediators that, combined with decreased ceramide apoptosis signaling molecules, may contribute to the tumor suppressor activity of Hint1.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Hepatology Research Group, Department of Clinical Research, University of Bern, Switzerland
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Juliette Martin
- Hepatology Research Group, Department of Clinical Research, University of Bern, Switzerland
| | - Olivier Maurhofer
- Hepatology Research Group, Department of Clinical Research, University of Bern, Switzerland
| | - Juliane Dorow
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jean-François Dufour
- Hepatology Research Group, Department of Clinical Research, University of Bern, Switzerland
| | - Jeffrey R Idle
- Hepatology Research Group, Department of Clinical Research, University of Bern, Switzerland Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
22
|
Alphonse G, Maalouf M, Battiston-Montagne P, Ardail D, Beuve M, Rousson R, Taucher-Scholz G, Fournier C, Rodriguez-Lafrasse C. p53-independent early and late apoptosis is mediated by ceramide after exposure of tumor cells to photon or carbon ion irradiation. BMC Cancer 2013; 13:151. [PMID: 23530619 PMCID: PMC3621616 DOI: 10.1186/1471-2407-13-151] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 03/12/2013] [Indexed: 01/22/2023] Open
Abstract
Background To determine whether ceramide is responsible for the induction of p53-independent early or late apoptosis in response to high- and low-Linear-Energy-Transfer (LET) irradiation. Methods Four cell lines displaying different radiosensitivities and p53-protein status were irradiated with photons or 33.4 or 184 keV/μm carbon ions. The kinetics of ceramide production was quantified by fluorescent microscopy or High-Performance-Liquid-Chromatogaphy and the sequence of events leading to apoptosis by flow cytometry. Results Regardless of the p53-status, both low and high-LET irradiation induced an early ceramide production in radiosensitive cells and late in the radioresistant. This production strongly correlated with the level of early apoptosis in radiosensitive cells and delayed apoptosis in the radioresistant ones, regardless of radiation quality, tumor type, radiosensitivity, or p53-status. Inhibition of caspase activity or ceramide production showed that, for both types of radiation, ceramide is essential for the initiation of early apoptosis in radiosensitive cells and late apoptosis following mitotic catastrophe in radioresistant cells. Conclusions Ceramide is a determining factor in the onset of early and late apoptosis after low and high-LET irradiation and is the mediator of the p53-independent-apoptotic pathway. We propose that ceramide is the molecular bridge between mitotic catastrophe and the commitment phase of delayed apoptosis in response to irradiation.
Collapse
|
23
|
Hage-Sleiman R, Esmerian MO, Kobeissy H, Dbaibo G. p53 and Ceramide as Collaborators in the Stress Response. Int J Mol Sci 2013; 14:4982-5012. [PMID: 23455468 PMCID: PMC3634419 DOI: 10.3390/ijms14034982] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 01/22/2013] [Accepted: 02/01/2013] [Indexed: 02/08/2023] Open
Abstract
The sphingolipid ceramide mediates various cellular processes in response to several extracellular stimuli. Some genotoxic stresses are able to induce p53-dependent ceramide accumulation leading to cell death. However, in other cases, in the absence of the tumor suppressor protein p53, apoptosis proceeds partly due to the activity of this "tumor suppressor lipid", ceramide. In the current review, we describe ceramide and its roles in signaling pathways such as cell cycle arrest, hypoxia, hyperoxia, cell death, and cancer. In a specific manner, we are elaborating on the role of ceramide in mitochondrial apoptotic cell death signaling. Furthermore, after highlighting the role and mechanism of action of p53 in apoptosis, we review the association of ceramide and p53 with respect to apoptosis. Strikingly, the hypothesis for a direct interaction between ceramide and p53 is less favored. Recent data suggest that ceramide can act either upstream or downstream of p53 protein through posttranscriptional regulation or through many potential mediators, respectively.
Collapse
Affiliation(s)
- Rouba Hage-Sleiman
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Infectious Diseases, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Riad El Solh, 1107 2020 Beirut, Lebanon; E-Mails: (M.O.E.); (G.D.)
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Riad El Solh, 1107 2020 Beirut, Lebanon; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +961-1-350-000 (ext. 4883)
| | - Maria O. Esmerian
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Infectious Diseases, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Riad El Solh, 1107 2020 Beirut, Lebanon; E-Mails: (M.O.E.); (G.D.)
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Riad El Solh, 1107 2020 Beirut, Lebanon; E-Mail:
| | - Hadile Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Riad El Solh, 1107 2020 Beirut, Lebanon; E-Mail:
| | - Ghassan Dbaibo
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Infectious Diseases, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Riad El Solh, 1107 2020 Beirut, Lebanon; E-Mails: (M.O.E.); (G.D.)
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, P.O. Box 11-0236 Riad El Solh, 1107 2020 Beirut, Lebanon; E-Mail:
| |
Collapse
|
24
|
Goldstein I, Rotter V. Regulation of lipid metabolism by p53 - fighting two villains with one sword. Trends Endocrinol Metab 2012; 23:567-75. [PMID: 22819212 DOI: 10.1016/j.tem.2012.06.007] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 06/19/2012] [Accepted: 06/20/2012] [Indexed: 01/01/2023]
Abstract
Both cellular and systemic metabolism of lipids are paramount for homeostasis, and their malfunction leads to devastating pathologies. Recently, exciting findings have linked the p53 tumor suppressor to the regulation of lipid metabolism. Here, we summarize these findings showing a clear role for p53 in enhancing lipid catabolism while inhibiting its anabolism. We also describe the multitude of genes regulated by p53 that participate in or regulate systemic lipid transport. From the compilation of available data a scenario is emerging in which p53 regulates genes involved in lipid metabolism - both in a cancer-preventive effort and, intriguingly, as a means to prevent atherosclerosis. Thus, by regulating lipid metabolism, p53 fights the two major causes of death worldwide - atherosclerosis and cancer.
Collapse
Affiliation(s)
- Ido Goldstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100 Israel
| | | |
Collapse
|
25
|
The control of the balance between ceramide and sphingosine-1-phosphate by sphingosine kinase: Oxidative stress and the seesaw of cell survival and death. Comp Biochem Physiol B Biochem Mol Biol 2012; 163:26-36. [DOI: 10.1016/j.cbpb.2012.05.006] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 05/09/2012] [Accepted: 05/12/2012] [Indexed: 12/19/2022]
|
26
|
Salminen A, Kauppinen A, Kaarniranta K. Emerging role of NF-κB signaling in the induction of senescence-associated secretory phenotype (SASP). Cell Signal 2012; 24:835-45. [PMID: 22182507 DOI: 10.1016/j.cellsig.2011.12.006] [Citation(s) in RCA: 480] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 12/04/2011] [Indexed: 11/17/2022]
Abstract
The major hallmark of cellular senescence is an irreversible cell cycle arrest and thus it is a potent tumor suppressor mechanism. Genotoxic insults, e.g. oxidative stress, are important inducers of the senescent phenotype which is characterized by an accumulation of senescence-associated heterochromatic foci (SAHF) and DNA segments with chromatin alterations reinforcing senescence (DNA-SCARS). Interestingly, senescent cells secrete pro-inflammatory factors and thus the condition has been called the senescence-associated secretory phenotype (SASP). Emerging data has revealed that NF-κB signaling is the major signaling pathway which stimulates the appearance of SASP. It is known that DNA damage provokes NF-κB signaling via a variety of signaling complexes containing NEMO protein, an NF-κB essential modifier, as well as via the activation of signaling pathways of p38MAPK and RIG-1, retinoic acid inducible gene-1. Genomic instability evoked by cellular stress triggers epigenetic changes, e.g. release of HMGB1 proteins which are also potent enhancers of inflammatory responses. Moreover, environmental stress and chronic inflammation can stimulate p38MAPK and ceramide signaling and induce cellular senescence with pro-inflammatory responses. On the other hand, two cyclin-dependent kinase inhibitors, p16INK4a and p14ARF, are effective inhibitors of NF-κB signaling. We will review in detail the signaling pathways which activate NF-κB signaling and trigger SASP in senescent cells.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FIN-70211 Kuopio, Finland.
| | | | | |
Collapse
|
27
|
Heffernan-Stroud LA, Helke KL, Jenkins RW, De Costa AM, Hannun YA, Obeid LM. Defining a role for sphingosine kinase 1 in p53-dependent tumors. Oncogene 2012; 31:1166-75. [PMID: 21765468 PMCID: PMC3278571 DOI: 10.1038/onc.2011.302] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2011] [Revised: 06/12/2011] [Accepted: 06/13/2011] [Indexed: 12/23/2022]
Abstract
p53 is a crucial tumor suppressor that is mutated or deleted in a majority of cancers. Exactly how p53 prevents tumor progression has proved elusive for many years; however, this information is crucial to define targets for chemotherapeutic development that can effectively restore p53 function. Bioactive sphingolipids have recently emerged as important regulators of proliferative, apoptotic and senescent cellular processes. In this study, we demonstrate that the enzyme sphingosine kinase 1 (SK1), a critical enzyme in the regulation of the key bioactive sphingolipids ceramide, sphingosine and sphingosine-1-phosphate (S1P), serves as a key downstream target for p53 action. Our results show that SK1 is proteolysed in response to genotoxic stress in a p53-dependent manner. p53 null mice display elevation of SK1 levels and a tumor-promoting dysregulation of bioactive sphingolipids in which the anti-growth sphingolipid ceramide is decreased and the pro-growth sphingolipid S1P is increased. Importantly, deletion of SK1 in p53 null mice completely abrogated thymic lymphomas in these mice and prolonged their life span by ~30%. Deletion of SK1 also significantly attenuated the formation of other cancers in p53 heterozygote mice. The mechanism of p53 tumor suppression by loss of SK1 is mediated by elevations of sphingosine and ceramide, which in turn were accompanied by increased expression of cell cycle inhibitors and tumor cell senescence. Thus, targeting SK1 may restore sphingolipid homeostasis in p53-dependent tumors and provide insights into novel therapeutic approaches to cancer.
Collapse
Affiliation(s)
- Linda A. Heffernan-Stroud
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Kristi L. Helke
- Department of Comparative Medicine/Lab Animal Resources, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Russell W. Jenkins
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Anna-Maria De Costa
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Yusuf A. Hannun
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29403, USA
| | - Lina M. Obeid
- Ralph H. Johnson VAMC, Charleston, SC
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC 29403, USA
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29403, USA
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29403, USA
| |
Collapse
|
28
|
de la Monte SM. Triangulated mal-signaling in Alzheimer's disease: roles of neurotoxic ceramides, ER stress, and insulin resistance reviewed. J Alzheimers Dis 2012; 30 Suppl 2:S231-49. [PMID: 22337830 PMCID: PMC4550324 DOI: 10.3233/jad-2012-111727] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ceramides are lipid signaling molecules that cause cytotoxicity and cell death mediated by insulin resistance, inflammation, and endoplasmic reticulum (ER) stress. However, insulin resistance dysregulates lipid metabolism, which promotes ceramide accumulation with attendant inflammation and ER stress. Herein, we discuss two major pathways, extrinsic and intrinsic, that converge and often overlap in propagating AD-type neurodegeneration via a triangulated mal-signaling network. First, we review evidence that systemic insulin resistance diseases linked to obesity, type 2 diabetes, and non-alcoholic steatohepatitis promote neurodegeneration. Mechanistically, we propose that toxic ceramides generated in extra-CNS tissues (e.g., liver) get released into peripheral blood, and subsequently transit across the blood-brain barrier into the brain where they induce brain insulin resistance, inflammation, and cell death (extrinsic pathway). Then we discuss the role of the intrinsic pathway of neurodegeneration which is mediated by endogenous or primary brain insulin/IGF resistance, and impairs neuronal and oligodendrocyte survival, energy metabolism, membrane integrity, cytoskeletal function, and AβPP-Aβ secretion. The end result is increased ER stress and ceramide generation, which exacerbate brain insulin resistance, cell death, myelin degeneration, and neuroinflammation. Altogether, the data suggest that the triangulated mal-signaling network mediated by toxic ceramides, ER stress, and insulin resistance should be targeted to disrupt positive feedback loops that drive the AD neurodegeneration cascade.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Pathology (Neuropathology), Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA. SuzanneDeLaMonte
| |
Collapse
|
29
|
Tuhvatulin A, Sysolyatina E, Scheblyakov D, Logunov D, Vasiliev M, Yurova M, Danilova M, Petrov O, Naroditsky B, Morfill G, Grigoriev A, Fortov V, Gintsburg A, Ermolaeva S. Non-thermal Plasma Causes p53-Dependent Apoptosis in Human Colon Carcinoma Cells. Acta Naturae 2012; 4:82-7. [PMID: 23150806 PMCID: PMC3491894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Non-thermal plasma (NTP) consists of a huge amount of biologically active particles, whereas its temperature is close to ambient. This combination allows one to use NTP as a perspective tool for solving different biomedical tasks, including antitumor therapy. The treatment of tumor cells with NTP caused dose-dependent effects, such as growth arrest and apoptosis. However, while the outcome of NTP treatment has been established, the molecular mechanisms of the interaction between NTP and eukaryotic cells have not been thoroughly studied thus far. In this work, the mechanisms and the type of death of human colon carcinoma HCT 116 cells upon application of non-thermal argon plasma were studied. The effect of NTP on the major stress-activated protein p53 was investigated. The results demonstrate that the viability of HCT116 cells upon plasma treatment is dependent on the functional p53 protein. NTP treatment caused an increase in the intracellular concentration of p53 and the induction of the p53-controlled regulon. The p53-dependent accumulation of active proapoptotic caspase-3 was shown in NTP-treated cells. The study was the first to demonstrate that treatment of human colon carcinoma cells with NTP results in p53-dependent apoptosis. The results obtained contribute to our understanding of the applicability of NTP in antitumor therapy.
Collapse
Affiliation(s)
- A.I. Tuhvatulin
- Gamaleya Research Institute of Epidemiology and Microbiology, Russian
Academy of Medical Sciences, 18, Gamaleya Str., 18, Moscow, Russia, 123098
| | - E.V. Sysolyatina
- Gamaleya Research Institute of Epidemiology and Microbiology, Russian
Academy of Medical Sciences, 18, Gamaleya Str., 18, Moscow, Russia, 123098
| | - D.V. Scheblyakov
- Gamaleya Research Institute of Epidemiology and Microbiology, Russian
Academy of Medical Sciences, 18, Gamaleya Str., 18, Moscow, Russia, 123098
| | - D.Yu. Logunov
- Gamaleya Research Institute of Epidemiology and Microbiology, Russian
Academy of Medical Sciences, 18, Gamaleya Str., 18, Moscow, Russia, 123098
| | - M.M. Vasiliev
- Joint Institute for High Temperatures, Russian Academy of Sciences,
Izhorskaya Str., 13/2, Moscow, Russia, 125412
| | - M.A. Yurova
- Gamaleya Research Institute of Epidemiology and Microbiology, Russian
Academy of Medical Sciences, 18, Gamaleya Str., 18, Moscow, Russia, 123098
| | - M.A. Danilova
- Gamaleya Research Institute of Epidemiology and Microbiology, Russian
Academy of Medical Sciences, 18, Gamaleya Str., 18, Moscow, Russia, 123098
| | - O.F. Petrov
- Joint Institute for High Temperatures, Russian Academy of Sciences,
Izhorskaya Str., 13/2, Moscow, Russia, 125412
| | - B.S. Naroditsky
- Gamaleya Research Institute of Epidemiology and Microbiology, Russian
Academy of Medical Sciences, 18, Gamaleya Str., 18, Moscow, Russia, 123098
| | - G.E. Morfill
- Max Planck Institute for Extraterrestrial Physics, Scheinerstrasse, 1,
Munich, Germany, 81679
| | - A.I. Grigoriev
- Institute for Biomedical Problems, Russian Academy of Sciences,
Horoshevskoe sh., 76 A, Moscow, Russia, 123007
| | - V.E. Fortov
- Joint Institute for High Temperatures, Russian Academy of Sciences,
Izhorskaya Str., 13/2, Moscow, Russia, 125412
| | - A.L. Gintsburg
- Gamaleya Research Institute of Epidemiology and Microbiology, Russian
Academy of Medical Sciences, 18, Gamaleya Str., 18, Moscow, Russia, 123098
| | - S.A. Ermolaeva
- Gamaleya Research Institute of Epidemiology and Microbiology, Russian
Academy of Medical Sciences, 18, Gamaleya Str., 18, Moscow, Russia, 123098
| |
Collapse
|