1
|
Geng Y, Lou J, Wu J, Tao Z, Yang N, Kuang J, Wu Y, Zhang J, Xiang L, Shi J, Cai Y, Wang X, Chen J, Xiao J, Zhou K. NEMO-Binding Domain/IKKγ Inhibitory Peptide Alleviates Neuronal Pyroptosis in Spinal Cord Injury by Inhibiting ASMase-Induced Lysosome Membrane Permeabilization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405759. [PMID: 39225315 PMCID: PMC11516130 DOI: 10.1002/advs.202405759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/16/2024] [Indexed: 09/04/2024]
Abstract
A short peptide termed NEMO-binding domain (NBD) peptide has an inhibitory effect on nuclear factor kappa-B (NF-κB). Despite its efficacy in inhibiting inflammatory responses, the precise neuroprotective mechanisms of NBD peptide in spinal cord injury (SCI) remain unclear. This study aims to determine whether the pyroptosis-related aspects involved in the neuroprotective effects of NBD peptide post-SCI.Using RNA sequencing, the molecular mechanisms of NBD peptide in SCI are explored. The evaluation of functional recovery is performed using the Basso mouse scale, Nissl staining, footprint analysis, Masson's trichrome staining, and HE staining. Western blotting, enzyme-linked immunosorbent assays, and immunofluorescence assays are used to examine pyroptosis, autophagy, lysosomal membrane permeabilization (LMP), acid sphingomyelinase (ASMase), and the NF-κB/p38-MAPK related signaling pathway.NBD peptide mitigated glial scar formation, reduced motor neuron death, and enhanced functional recovery in SCI mice. Additionally, NBD peptide inhibits pyroptosis, ameliorate LMP-induced autophagy flux disorder in neuron post-SCI. Mechanistically, NBD peptide alleviates LMP and subsequently enhances autophagy by inhibiting ASMase through the NF-κB/p38-MAPK/Elk-1/Egr-1 signaling cascade, thereby mitigating neuronal death. NBD peptide contributes to functional restoration by suppressing ASMase-mediated LMP and autophagy depression, and inhibiting pyroptosis in neuron following SCI, which may have potential clinical application value.
Collapse
Affiliation(s)
- Yibo Geng
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Junsheng Lou
- Department of Orthopedic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003China
| | - Junnan Wu
- Department of PharmacyThe Quzhou Affiliated Hospital of Wenzhou Medical UniversityQuzhou People's HospitalQuzhou324000China
| | - Zhichao Tao
- Renji College of Wenzhou Medical UniversityWenzhou325027China
| | - Ningning Yang
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Jiaxuan Kuang
- Cixi Biomedical Research InstituteWenzhou Medical UniversityNingbo315300China
| | - Yuzhe Wu
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Jiacheng Zhang
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Linyi Xiang
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Jingwei Shi
- Cixi Biomedical Research InstituteWenzhou Medical UniversityNingbo315300China
| | - Yuepiao Cai
- Cixi Biomedical Research InstituteWenzhou Medical UniversityNingbo315300China
- Molecular Pharmacology Research CenterSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhou325027China
| | - Xiangyang Wang
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Jiaoxiang Chen
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
| | - Jian Xiao
- Cixi Biomedical Research InstituteWenzhou Medical UniversityNingbo315300China
- Molecular Pharmacology Research CenterSchool of Pharmaceutical ScienceWenzhou Medical UniversityWenzhou325027China
| | - Kailiang Zhou
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhou325027China
- Zhejiang Provincial Key Laboratory of OrthopaedicsWenzhou325027China
- Cixi Biomedical Research InstituteWenzhou Medical UniversityNingbo315300China
| |
Collapse
|
2
|
Lopuszynski J, Wang J, Zahid M. Beyond Transduction: Anti-Inflammatory Effects of Cell Penetrating Peptides. Molecules 2024; 29:4088. [PMID: 39274936 PMCID: PMC11397606 DOI: 10.3390/molecules29174088] [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: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
One of the bottlenecks to bringing new therapies to the clinic has been a lack of vectors for delivering novel therapeutics in a targeted manner. Cell penetrating peptides (CPPs) have received a lot of attention and have been the subject of numerous developments since their identification nearly three decades ago. Known for their transduction abilities, they have generally been considered inert vectors. In this review, we present a schema for their classification, highlight what is known about their mechanism of transduction, and outline the existing literature as well as our own experience, vis a vis the intrinsic anti-inflammatory properties that certain CPPs exhibit. Given the inflammatory responses associated with viral vectors, CPPs represent a viable alternative to such vectors; furthermore, the anti-inflammatory properties of CPPs, mostly through inhibition of the NF-κB pathway, are encouraging. Much more work in relevant animal models, toxicity studies in large animal models, and ultimately human trials are needed before their potential is fully realized.
Collapse
Affiliation(s)
| | | | - Maliha Zahid
- Department of Cardiovascular Medicine, Guggenheim Gu 9-01B, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
3
|
Manabe Y, Takagi-Hayashi S, Mohri S, Sugawara T. Intestinal Absorption and Anti-Inflammatory Effects of Siphonein, a Siphonaxanthin Fatty Acid Ester from Green Algae. J Nutr Sci Vitaminol (Tokyo) 2023; 69:62-70. [PMID: 36858542 DOI: 10.3177/jnsv.69.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Siphonein is a C19 acylated siphonaxanthin found in some edible green algae (e.g., Codium fragile and Caulerpa lentillifera). Although the content of siphonein in these green algae is similar to or higher than that of siphonaxanthin, studies of health-related biological activity of siphonein are much less than those of siphonaxanthin. Given the difference in the position of the acyl chain, one cannot infer intestinal absorption of siphonein from other general carotenoid fatty acid esters. In this study, we first investigated the intestinal absorption of siphonein using mouse and cell culture models. A small amount of siphonein was detected in the plasma of treated mice, and its concentration was higher than that of siphonaxanthin (i.e., the hydrolyzed product of ingested siphonein) from 1 to 6 h after administration. Pharmacological inhibition tests with differentiated Caco-2 cells showed that Nieman-Pick C1-like 1-mediated facilitated diffusion was involved in the cellular uptake of siphonein. These results indicate that, unlike general carotenoid fatty acid esters, siphonein can be absorbed without hydrolysis. We also evaluated the anti-inflammatory effect of siphonein in differentiated Caco-2 cells. Siphonein pretreatment modulated lipopolysaccharide-induced cellular lipidome alterations and suppressed mRNA expression of proinflammatory chemokines, CXCL8 protein release, and activation of NF-κB. This study provides new insights into the absorption processes of carotenoids and shows the anti-inflammatory effect of siphonein for the first time.
Collapse
Affiliation(s)
- Yuki Manabe
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University
| | | | - Shinsuke Mohri
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University
| | - Tatsuya Sugawara
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University
| |
Collapse
|
4
|
Zhang J, Zhou N, Wang Y, Liu T, Cao Y, Feng W, Zheng X. Protective effects of Descurainia sophia seeds extract and its fractions on pulmonary edema by untargeted urine and serum metabolomics strategy. Front Pharmacol 2023; 14:1080962. [PMID: 36865914 PMCID: PMC9971919 DOI: 10.3389/fphar.2023.1080962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
Background: Descurainia sophia seeds (DS) is a herbal medicine in traditional Chinese medicine (TCM) for treating lung diseases. We aimed to evaluate the therapeutic effect of DS and five of its fractions upon pulmonary edema (PE) through metabolomics analysis (MA) of urine and serum samples of rats. Methods: A PE model was established by intrathoracic injection of carrageenan. Rats were pretreated with DS extract or its five fractions (polysaccharides (DS-Pol); oligosaccharides (DS-Oli); flavonoid glycosides (DS-FG); flavonoid aglycone (DS-FA); fat oil fraction (DS-FO)) for seven consecutive days. Forty-eight hours after carrageenan injection, lung tissues were subjected to histopathology. MA of urine and serum was done by ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry, respectively. Principal component analysis and orthogonal partial least squares-discriminant analysis were operated for the MA of rats and potential biomarkers related to treatment. Heatmaps and metabolic networks were constructed to explore how DS and its five fractions act against PE. Results: DS and its five fractions could all attenuate pathologic lung injury to different degrees, and DS-Oli, DS-FG, and DS-FO had a more potent effect compared with DS-Pol and DS-FA. DS-Oli, DS-FG, DS-FA, and DS-FO could regulate the metabolic profiles of PE rats, but DS-Pol was less potent. According to MA, the five fractions could improve PE to some degree due to their anti-inflammatory, immunoregulatory, and renoprotective activities by mediating the metabolism of taurine, tryptophan, and arachidonic acid. However, DS-Oli, DS-FG, and DS-FO had more important roles in edema-fluid reabsorption, and reduction of vascular leakage through regulating the metabolism of phenylalanine, sphingolipid and bile acid. Finally, heatmaps and hierarchical clustering analysis indicated DS-Oli, DS-FG, and DS-FO to be more efficacious than DS-Pol or DS-FA against PE. The five fractions of DS had a synergistic effect on PE from different aspects, thereby constituting the entire efficacy of DS. DS-Oli, DS-FG, or DS-FO could be used as an alternative to DS. Conclusion: MA combined with use of DS and its fractions provided novel insights into the mechanism of action of TCM.
Collapse
Affiliation(s)
- Jinying Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ning Zhou
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China,The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China
| | - Yongxiang Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tong Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yumin Cao
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Weisheng Feng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China,The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China,Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan, Education Ministry of P.R, Zhengzhou, China,*Correspondence: Weisheng Feng, ; Xiaoke Zheng,
| | - Xiaoke Zheng
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China,The Engineering and Technology Center for Chinese Medicine Development of Henan Province, Zhengzhou, China,Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan, Education Ministry of P.R, Zhengzhou, China,*Correspondence: Weisheng Feng, ; Xiaoke Zheng,
| |
Collapse
|
5
|
Tang L, Zhang S, Zhang M, Wang P, Liang G, Gao X. Analysis of protective effects of Rosa Roxburghii Tratt fruit polyphenols on lipopolysaccharide-induced acute lung injury through network pharmacology and metabolomics. Food Sci Nutr 2022; 10:4258-4269. [PMID: 36514748 PMCID: PMC9731534 DOI: 10.1002/fsn3.3019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/20/2022] [Accepted: 07/24/2022] [Indexed: 12/16/2022] Open
Abstract
Acute lung injury (ALI) is a respiratory disease with high morbidity and mortality rates and is the primary cause of death in children and the elderly around the world. The use of Chinese foods in the complementary and alternative treatment of ALI has attracted more and more attention. This study aimed to explore the anti-ALI activity of Chinese functional foods Rosa roxburghii Tratt fruit polyphenols (RRTP). RRTP was administered to lipopolysaccharide-induced ALI mice, and its protective effects were comprehensively evaluated by lung histopathological examination, wet/dry (W/D) ratio, and cytokine production. Metabolomics analysis was used to identify the differential metabolites and metabolic pathways in plasma, and molecular docking and systemic biology-based network pharmacology assay were performed to explore the active components and potential therapeutic targets. The results indicated that RRTP significantly attenuated the severity of pathological changes and pulmonary capillary permeability. Furthermore, RRTP limited the increase in tumor necrosis factor alpha (TNF-α), interleukin 1β (IL-1β), and interleukin 6 (IL-6) levels and the decrease in interleukin 10 (IL-10) levels in ALI mice. Metabolomics studies revealed that RRTP markedly affected 19 different metabolites, three amino acid metabolism pathways, and sphingolipid metabolism. Moreover, network pharmacology identified AKT1 (AKT serine/threonine kinase 1), TP53, IL-6, VEGFA (vascular endothelial growth factor A), and TNF (tumor necrosis factor) as the most promising target proteins, while quercetin, luteolin, and kaempferol were the core active components of RRTP. This study investigated the complex mechanisms of RRTP against ALI for the first time, and provided a foundation for the application of RRTP as a functional food, facilitating the research of nutritional food additives for the adjuvant treatment of ALI.
Collapse
Affiliation(s)
- Li Tang
- School of Basic Medical Sciences & State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of EducationGuizhou Medical UniversityGuiyangChina
- School of Ethnic MedicineGuizhou Minzu UniversityGuiyangChina
| | - Shuo Zhang
- School of Basic Medical Sciences & State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of EducationGuizhou Medical UniversityGuiyangChina
| | - Min Zhang
- School of Basic Medical Sciences & State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of EducationGuizhou Medical UniversityGuiyangChina
| | - Peng‐Jiao Wang
- School of Basic Medical Sciences & State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of EducationGuizhou Medical UniversityGuiyangChina
| | - Gui‐You Liang
- School of Basic Medical Sciences & State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Translational Medicine Research CenterGuizhou Medical UniversityGuiyangChina
| | - Xiu‐Li Gao
- School of Basic Medical Sciences & State Key Laboratory of Functions and Applications of Medicinal PlantsGuizhou Medical UniversityGuiyangChina
- Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of EducationGuizhou Medical UniversityGuiyangChina
- Translational Medicine Research CenterGuizhou Medical UniversityGuiyangChina
| |
Collapse
|
6
|
Jiang Y, He X, Simonaro CM, Yi B, Schuchman EH. Acid Ceramidase Protects Against Hepatic Ischemia/Reperfusion Injury by Modulating Sphingolipid Metabolism and Reducing Inflammation and Oxidative Stress. Front Cell Dev Biol 2021; 9:633657. [PMID: 34026750 PMCID: PMC8134688 DOI: 10.3389/fcell.2021.633657] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/12/2021] [Indexed: 11/30/2022] Open
Abstract
Ceramide is a bioactive signaling lipid involved in the pathogenesis of numerous diseases. It also plays an important role in ischemia reperfusion (IR) injury via activation of inflammatory/oxidative stress-stimulated signaling pathways, resulting in tissue damage. Acid ceramidase is a lipid hydrolase that modulates the levels of ceramide, and as such has a potential therapeutic role in many human diseases where ceramide has been implicated. Here we investigated the therapeutic potential of recombinant acid ceramidase in a murine model of hepatic IR injury. Serum ALT, AST, and LDH activities, as well as oxidative stress (MDA) and inflammatory (MCP-1) markers, were increased in mice subjected to IR compared to a sham group. In contrast, these elevations were significantly lower in an IR group pretreated with a single injection of acid ceramidase. Histological examination by two different assessment criteria also revealed that acid ceramidase pretreatment alleviated IR-induced hepatocyte damage, including reduced evidence of cell death and necrosis. In addition, elevated ceramide and sphingosine levels were observed in the IR group compared to sham, and were markedly reduced when pretreated with acid ceramidase. In contrast, the levels of the protective signaling lipid, sphingosine-1-phosphate (S1P), were reduced following IR and elevated in response to acid ceramidase pretreatment. These changes in sphingolipid levels could be correlated with changes in the activities of several sphingolipid-metabolizing enzymes. Overall, these results indicated that sphingolipid changes were an important pathologic component of hepatic IR injury, and that acid ceramidase administration ameliorated these lipid changes and other downstream pathologic changes.
Collapse
Affiliation(s)
- Yuan Jiang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China.,Department of Clinical Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Xingxuan He
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Calogera M Simonaro
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Bin Yi
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Edward H Schuchman
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
7
|
Kopincova J, Mikolka P, Kolomaznik M, Kosutova P, Calkovska A, Mokra D. Selective inhibition of NF-kappaB and surfactant therapy in experimental meconium-induced lung injury. Physiol Res 2018; 66:S227-S236. [PMID: 28937237 DOI: 10.33549/physiolres.933678] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Meconium aspiration syndrome (MAS) in newborns is characterized mainly by respiratory failure due to surfactant dysfunction and inflammation. Previous meta-analyses did not prove any effect of exogenous surfactant treatment nor glucocorticoid administration on final outcome of children with MAS despite oxygenation improvement. As we supposed there is the need to intervene in both these fields simultaneously, we evaluated therapeutic effect of combination of exogenous surfactant and selective inhibitor of NF-kappaB (IKK-NBD peptide). Young New Zealand rabbits were instilled by meconium suspension and treated by surfactant alone or surfactant in combination with IKK-NBD, and oxygen-ventilated for 5 h. PaO(2)/FiO(2), oxygenation index, oxygen saturation and ventilation efficiency index were evaluated every hour; post mortem, total and differential leukocyte counts were investigated in bronchoalveolar lavage fluid (BALF) and inflammatory, oxidative and apoptotic markers were assessed in lung tissue homogenates. Exogenous surfactant combined with IKK-NBD improved oxygenation, reduced neutrophil count in BALF and levels of IL-1beta, IL-6, p38 MAPK and caspase 3 in comparison with surfactant-only therapy. It seems that inhibition of inflammation may be strong supporting factor in surfactant treatment of MAS.
Collapse
Affiliation(s)
- J Kopincova
- Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia.
| | | | | | | | | | | |
Collapse
|
8
|
The role of sphingolipid metabolism disruption on lipopolysaccharide-induced lung injury in mice. Pulm Pharmacol Ther 2018; 50:100-110. [DOI: 10.1016/j.pupt.2018.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/08/2018] [Accepted: 04/21/2018] [Indexed: 02/01/2023]
|
9
|
Wang K, Xu R, Snider AJ, Schrandt J, Li Y, Bialkowska AB, Li M, Zhou J, Hannun YA, Obeid LM, Yang VW, Mao C. Alkaline ceramidase 3 deficiency aggravates colitis and colitis-associated tumorigenesis in mice by hyperactivating the innate immune system. Cell Death Dis 2016; 7:e2124. [PMID: 26938296 PMCID: PMC4823937 DOI: 10.1038/cddis.2016.36] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/13/2016] [Accepted: 01/19/2016] [Indexed: 12/16/2022]
Abstract
Increasing studies suggest that ceramides differing in acyl chain length and/or degree of unsaturation have distinct roles in mediating biological responses. However, still much remains unclear about regulation and role of distinct ceramide species in the immune response. Here, we demonstrate that alkaline ceramidase 3 (Acer3) mediates the immune response by regulating the levels of C18:1-ceramide in cells of the innate immune system and that Acer3 deficiency aggravates colitis in a murine model by augmenting the expression of pro-inflammatory cytokines in myeloid and colonic epithelial cells (CECs). According to the NCBI Gene Expression Omnibus (GEO) database, ACER3 is downregulated in immune cells in response to lipopolysaccharides (LPS), a potent inducer of the innate immune response. Consistent with these data, we demonstrated that LPS downregulated both Acer3 mRNA levels and its enzymatic activity while elevating C(18:1)-ceramide, a substrate of Acer3, in murine immune cells or CECs. Knocking out Acer3 enhanced the elevation of C(18:1)-ceramide and the expression of pro-inflammatory cytokines in immune cells and CECs in response to LPS challenge. Similar to Acer3 knockout, treatment with C(18:1)-ceramide, but not C18:0-ceramide, potentiated LPS-induced expression of pro-inflammatory cytokines in immune cells. In the mouse model of dextran sulfate sodium-induced colitis, Acer3 deficiency augmented colitis-associated elevation of colonic C(18:1)-ceramide and pro-inflammatory cytokines. Acer3 deficiency aggravated diarrhea, rectal bleeding, weight loss and mortality. Pathological analyses revealed that Acer3 deficiency augmented colonic shortening, immune cell infiltration, colonic epithelial damage and systemic inflammation. Acer3 deficiency also aggravated colonic dysplasia in a mouse model of colitis-associated colorectal cancer. Taken together, these results suggest that Acer3 has an important anti-inflammatory role by suppressing cellular or tissue C(18:1)-ceramide, a potent pro-inflammatory bioactive lipid and that dysregulation of ACER3 and C(18:1)-ceramide may contribute to the pathogenesis of inflammatory diseases including cancer.
Collapse
Affiliation(s)
- K Wang
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - R Xu
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| | - A J Snider
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
- Northport Veterans Affairs Medical Center, Northport, NY, USA
| | - J Schrandt
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
| | - Y Li
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| | - A B Bialkowska
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
| | - M Li
- Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - J Zhou
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Y A Hannun
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| | - L M Obeid
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
- Northport Veterans Affairs Medical Center, Northport, NY, USA
| | - V W Yang
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
| | - C Mao
- Department of Medicine, State University of New York at Stony Brook University, Stony Brook, NY, USA
- Stony Brook Cancer Center, Stony Brook, NY, USA
| |
Collapse
|
10
|
Petrache I, Berdyshev EV. Ceramide Signaling and Metabolism in Pathophysiological States of the Lung. Annu Rev Physiol 2015; 78:463-80. [PMID: 26667073 DOI: 10.1146/annurev-physiol-021115-105221] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Following the discovery of ceramide as the central signaling and metabolic relay among sphingolipids, studies of its involvement in lung health and pathophysiology have exponentially increased. In this review, we highlight key studies in the context of recent progress in metabolomics and translational research methodologies. Evidence points toward an important role for the ceramide/sphingosine-1-phosphate rheostat in maintaining lung cell survival, vascular barrier function, and proper host response to airway microbial infections. Sphingosine kinase 1 has emerged as an important determinant of sphingosine-1-phosphate lung levels, which, when aberrantly high, contribute to lung fibrosis, maladaptive vascular remodeling, and allergic asthma. New sphingolipid metabolites have been discovered as potential biomarkers of several lung diseases. Although multiple acute and chronic lung pathological conditions involve perturbations in sphingolipid signaling and metabolism, there are specific patterns, unique sphingolipid species, enzymes, metabolites, and receptors, which have emerged that deepen our understanding of lung pathophysiology and inform the development of new therapies for lung diseases.
Collapse
Affiliation(s)
- Irina Petrache
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, National Jewish Health, Denver, Colorado 80206; ,
| | - Evgeny V Berdyshev
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, National Jewish Health, Denver, Colorado 80206; ,
| |
Collapse
|
11
|
Gonzalo S, Valero MS, Martínez de Salinas F, Vergara C, Arruebo MP, Plaza MÁ, Murillo MD, Grasa L. Roles of Toll-Like Receptor 4, IκB Kinase, and the Proteasome in the Intestinal Alterations Caused by Sepsis. Dig Dis Sci 2015; 60:1223-31. [PMID: 25371155 DOI: 10.1007/s10620-014-3418-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 10/29/2014] [Indexed: 01/03/2023]
Abstract
BACKGROUND Lipopolysaccharide decreases intestinal contractility and induces the production of cytokines, which play an important role in the pathogenesis of sepsis. AIM The objective of the present study was to examine the role of Toll-like receptor 4, IκB kinase, and the proteasome in the intestinal alterations induced by lipopolysaccharide. METHODS Sepsis was induced in rabbits by intravenous injection of lipopolysaccharide. Contractility studies of rabbit duodenum were performed in an organ bath. Expressions of interleukin-1β, interleukin-6, interleukin-8, interleukin-10, IκB kinase-α, IκB kinase-β, IκB kinase-γ, and the proteasome mRNA were determined by RT-PCR on rabbit duodenum. RESULTS Neomycin and polymyxin B (Toll-like receptor 4 inhibitors), IKK NBD peptide (IκB kinase complex inhibitor), and MG-132 (proteasome inhibitor) blocked partially the effects of lipopolysaccharide on the acetylcholine-, prostaglandin E2-, substance P-, and KCl-induced contractions in the longitudinal and circular smooth muscle of rabbit duodenum. Lipopolysaccharide increased the mRNA expression of interleukin-6 and interleukin-8 in duodenal tissue, and this effect was partly reversed by neomycin, polymyxin B, IKK NBD peptide, and MG-132. IκB kinase-α, IκB kinase-β, IκB kinase-γ, and the proteasome mRNA expressions was not affected by lipopolysaccharide treatment. CONCLUSIONS Toll-like receptor 4, the IκB kinase complex, and the proteasome could be therapeutic targets in the treatment of sepsis symptoms in the intestine.
Collapse
Affiliation(s)
- Sergio Gonzalo
- Department of Pharmacology and Physiology, Faculty of Veterinary Medicine, University of Zaragoza, c/ Miguel Servet 177, 50013, Saragossa, Spain,
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Yu XH, Zheng XL, Tang CK. Nuclear Factor-κB Activation as a Pathological Mechanism of Lipid Metabolism and Atherosclerosis. Adv Clin Chem 2015; 70:1-30. [PMID: 26231484 DOI: 10.1016/bs.acc.2015.03.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall with lipid-laden lesions, involving a complex interaction between multiple different cell types and cytokine networks. Inflammatory responses mark all stages of atherogenesis: from lipid accumulation in the intima to plaque formation and eventual rupture. One of the most important regulators of inflammation is the transcription factor nuclear factor-κB (NF-κB), which is activated through the canonical and noncanonical pathways in response to various stimuli. NF-κB has long been regarded as a proatherogenic factor, because it is implicated in multiple pathological processes during atherogenesis, including foam cell formation, vascular inflammation, proliferation of vascular smooth muscle cells, arterial calcification, and plaque progression. In contrast, inhibition of NF-κB signaling has been shown to protect against atherosclerosis. This chapter aims to discuss recent progress on the roles of NF-κB in lipid metabolism and atherosclerosis and also to highlight its potential therapeutic benefits.
Collapse
Affiliation(s)
- Xiao-Hua Yu
- Key Laboratory for Atherosclerology of Hunan Province, Molecular Target New Drug Discovery and Cooperative Innovation Center of Hunan Province, Life Science Research Center, University of South China, Hengyang, PR China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Health Sciences Center, Calgary, Alberta, Canada
| | - Chao-Ke Tang
- Key Laboratory for Atherosclerology of Hunan Province, Molecular Target New Drug Discovery and Cooperative Innovation Center of Hunan Province, Life Science Research Center, University of South China, Hengyang, PR China.
| |
Collapse
|
13
|
Swarnkar G, Abu-Amer Y. Regulation of NF-κB signaling in osteoclasts and myeloid progenitors. Methods Mol Biol 2015; 1280:527-42. [PMID: 25736770 DOI: 10.1007/978-1-4939-2422-6_31] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is crucial for immune responses and skeletal development. Work in recent years has shown that various members of the NF-κB family are viable targets to regulate activity and survival of bone cells and hence bone metabolism. In this regard, deletion of upstream kinases or distal NF-κB subunits resulted with bone deformities. Thus, it has become increasingly apparent that detailed investigation of NF-κB in bone cells may provide opportunities to design new therapeutic modalities. In this chapter we present modified methodology describing efficient approaches to regulate the NF-κB pathway in vitro and in vivo to assess its function in bone cells and tissues.
Collapse
Affiliation(s)
- Gaurav Swarnkar
- Department of Orthopedic Surgery-Research, Washington University School of Medicine, 660 S. Euclid Ave, Campus Box 8233, Saint Louis, MO, 63110, USA
| | | |
Collapse
|
14
|
Zhou L, Yeo AT, Ballarano C, Weber U, Allen KN, Gilmore TD, Whitty A. Disulfide-mediated stabilization of the IκB kinase binding domain of NF-κB essential modulator (NEMO). Biochemistry 2014; 53:7929-44. [PMID: 25400026 PMCID: PMC4278678 DOI: 10.1021/bi500920n] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Human NEMO (NF-κB
essential modulator) is a 419 residue scaffolding
protein that, together with catalytic subunits IKKα and IKKβ,
forms the IκB kinase (IKK) complex, a key regulator of NF-κB
pathway signaling. NEMO is an elongated homodimer comprising mostly
α-helix. It has been shown that a NEMO fragment spanning residues
44–111, which contains the IKKα/β binding site,
is structurally disordered in the absence of bound IKKβ. Herein
we show that enforcing dimerization of NEMO1–120 or NEMO44–111 constructs through introduction
of one or two interchain disulfide bonds, through oxidation of the
native Cys54 residue and/or at position 107 through a Leu107Cys mutation,
induces a stable α-helical coiled-coil structure that is preorganized
to bind IKKβ with high affinity. Chemical and thermal denaturation
studies showed that, in the context of a covalent dimer, the ordered
structure was stabilized relative to the denatured state by up to
3 kcal/mol. A full-length NEMO-L107C protein formed covalent dimers
upon treatment of mammalian cells with H2O2.
Furthermore, NEMO-L107C bound endogenous IKKβ in A293T cells,
reconstituted TNF-induced NF-κB signaling in NEMO-deficient
cells, and interacted with TRAF6. Our results indicate that the IKKβ
binding domain of NEMO possesses an ordered structure in the unbound
state, provided that it is constrained within a dimer as is the case
in the constitutively dimeric full-length NEMO protein. The stability
of the NEMO coiled coil is maintained by strong interhelix interactions
in the region centered on residue 54. The disulfide-linked constructs
we describe herein may be useful for crystallization of NEMO’s
IKKβ binding domain in the absence of bound IKKβ, thereby
facilitating the structural characterization of small-molecule inhibitors.
Collapse
Affiliation(s)
- Li Zhou
- Department of Chemistry and ‡Department of Biology, Boston University , Boston, Massachusetts 02215, United States
| | | | | | | | | | | | | |
Collapse
|
15
|
Sun D, Zhou M, Ying X, Cheng B, Han Y, Nie Y, Hou Y, Bai G. Identification of nuclear factor-κB inhibitors in the folk herb Rhizoma Menispermi via bioactivity-based ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry analysis. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:356. [PMID: 25252930 PMCID: PMC4195958 DOI: 10.1186/1472-6882-14-356] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/19/2014] [Indexed: 12/28/2022]
Abstract
BACKGROUND Rhizoma Menispermi (RM) is the dried root of Menispermum dauricum DC, which is traditionally used to treat swelling and pain for sore throat, enteritis and rheumatic arthralgia in the clinic, but its bioactive compounds remain unclear. METHODS In this study, RM extract was administered orally to ICR mice followed by challenging with an intratracheal Pseudomonas aeruginosa suspension. Then mortality, histological features of lung, and inflammatory cytokines were evaluated. RM treatment significantly ameliorated Pseudomonas aeruginosa-induced acute lung inflammation and reduced levels of inflammatory mediators. To screen for potential anti-inflammatory constituents of the RM extract, a simple and rapid method based on ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry (UPLC-Q/TOF MS) coupled with a luciferase reporter assay system to detect nuclear factor-κB (NF-κB) activity was established. RESULTS Using this system, seven potential NF-κB inhibitors were detected, including sinomenine, norsinoacutin, N-norsinoacutin-β-D-glucopyranoside, 6-O-methyl-laudanosoline-13-O-glucopyranoside, magnoflorine, laurifloline and dauricinoline. Furthermore, IL-6 and IL-8 assays confirmed the anti-inflammatory effects of these potential NF-κB inhibitors, in which norsinoacutin, 6-O-methyl-laudanosoline-13-O-glucopyranoside laurifloline, dauricinoline and N-norsinoacutin-β-D-glucopyranoside were revealed as new NF-κB inhibitors. CONCLUSION This method of UPLC-Q/TOF coupled with the luciferase reporter assay system was initially applied to the study of RM and was demonstrated to represent a simple, rapid and practical approach to screen for anti-inflammatory compounds. This study provided useful results for further investigation on the anti-inflammatory mechanism of RM.
Collapse
Affiliation(s)
- Dan Sun
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071 China
| | - Mengge Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071 China
| | - Xuhui Ying
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071 China
| | - Binfeng Cheng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071 China
| | - Yanqi Han
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071 China
| | - Yan Nie
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071 China
| | - Yuanyuan Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071 China
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300071 China
| |
Collapse
|
16
|
Improvement of ventilation-induced lung injury in a rodent model by inhibition of inhibitory κB kinase. J Trauma Acute Care Surg 2014; 76:1417-24. [DOI: 10.1097/ta.0000000000000229] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Habineza Ndikuyeze G, Gaurnier-Hausser A, Patel R, Baldwin AS, May MJ, Flood P, Krick E, Propert KJ, Mason NJ. A phase I clinical trial of systemically delivered NEMO binding domain peptide in dogs with spontaneous activated B-cell like diffuse large B-cell lymphoma. PLoS One 2014; 9:e95404. [PMID: 24798348 PMCID: PMC4010398 DOI: 10.1371/journal.pone.0095404] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/25/2014] [Indexed: 01/03/2023] Open
Abstract
Activated B-Cell (ABC) Diffuse Large B-Cell Lymphoma (DLBCL) is a common, aggressive and poorly chemoresponsive subtype of DLBCL, characterized by constitutive canonical NF-κB signaling. Inhibition of NF-κB signaling leads to apoptosis of ABC-DLBCL cell lines, suggesting targeted disruption of this pathway may have therapeutic relevance. The selective IKK inhibitor, NEMO Binding Domain (NBD) peptide effectively blocks constitutive NF-κB activity and induces apoptosis in ABC-DLBCL cells in vitro. Here we used a comparative approach to determine the safety and efficacy of systemic NBD peptide to inhibit constitutive NF-κB signaling in privately owned dogs with spontaneous newly diagnosed or relapsed ABC-like DLBCL. Malignant lymph nodes biopsies were taken before and twenty-four hours after peptide administration to determine biological effects. Intravenous administration of <2 mg/kg NBD peptide was safe and inhibited constitutive canonical NF-κB activity in 6/10 dogs. Reductions in mitotic index and Cyclin D expression also occurred in a subset of dogs 24 hours post peptide and in 3 dogs marked, therapeutically beneficial histopathological changes were identified. Mild, grade 1 toxicities were noted in 3 dogs at the time of peptide administration and one dog developed transient subclinical hepatopathy. Long term toxicities were not identified. Pharmacokinetic data suggested rapid uptake of peptide into tissues. No significant hematological or biochemical toxicities were identified. Overall the results from this phase I study indicate that systemic administration of NBD peptide is safe and effectively blocks constitutive NF-κB signaling and reduces malignant B cell proliferation in a subset of dogs with ABC-like DLBCL. These results have potential translational relevance for human ABC-DLBCL.
Collapse
Affiliation(s)
- Georges Habineza Ndikuyeze
- Division of Hematology/Oncology, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Anita Gaurnier-Hausser
- Office of Professional Studies in the Health Sciences, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Reema Patel
- Antech Diagnostics, New Hyde Park, New York, United States of America
| | - Albert S. Baldwin
- TheraLogics, Inc., Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center and Department of Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Michael J. May
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Patrick Flood
- 7-020G Katz Centre for Pharmacy and Health Research, The University of Alberta, Edmonton, Alberta, Canada
| | - Erika Krick
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kathleen J. Propert
- Department of Biostatistics and Epidemiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Nicola J. Mason
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
18
|
Caretti A, Bragonzi A, Facchini M, De Fino I, Riva C, Gasco P, Musicanti C, Casas J, Fabriàs G, Ghidoni R, Signorelli P. Anti-inflammatory action of lipid nanocarrier-delivered myriocin: therapeutic potential in cystic fibrosis. Biochim Biophys Acta Gen Subj 2013; 1840:586-94. [PMID: 24141140 DOI: 10.1016/j.bbagen.2013.10.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/07/2013] [Accepted: 10/10/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND Sphingolipids take part in immune response and can initiate and/or sustain inflammation. Various inflammatory diseases have been associated with increased ceramide content, and pharmacological reduction of ceramide diminishes inflammation damage in vivo. Inflammation and susceptibility to microbial infection are two elements in a vicious circle. Recently, sphingolipid metabolism inhibitors were used to reduce infection. Cystic fibrosis (CF) is characterized by a hyper-inflammation and an excessive innate immune response, which fails to evolve into adaptive immunity and to eradicate infection. Chronic infections result in lung damage and patient morbidity. Notably, ceramide content in mucosa airways is higher in CF mouse models and in patients than in control mice or healthy subjects. METHODS The therapeutic potential of myriocin, an inhibitor of the sphingolipid de novo synthesis rate limiting enzyme (Serine Palmitoyl Transferase, SPT),was investigated in CF cells and mice models. RESULTS We treated CF human respiratory epithelial cells with myriocin, This treatment resulted in reduced basal, as well as TNFα-stimulated, inflammation. In turn, TNFα induced an increase in SPT in these cells, linking de novo synthesis of ceramide to inflammation. Furthermore, myriocin-loaded nanocarrier, injected intratrachea prior to P. aeruginosa challenge, enabled a significant reduction of lung infection and reduced inflammation. CONCLUSIONS The presented data suggest that de novo ceramide synthesis is constitutively enhanced in CF mucosa and that it can be envisaged as pharmacological target for modulating inflammation and restoring effective innate immunity against acute infection. GENERAL SIGNIFICANCE Myriocin stands as a powerful immunomodulatory agent for inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Anna Caretti
- Department of Health Sciences, University of Milan, San Paolo Hospital, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Acute lung injury is a life-threatening disease that is characterized by pulmonary inflammation, loss of barrier functions, and hypoxemia. Sphingolipids are critically involved in the disease process that they can both expedite and extenuate: They expedite inflammation by promoting chemotaxis (neutral sphingomyelinase), increased endothelial permeability (acid sphingomyelinase, S1P3-receptors), increased epithelial permeability (S1P2- and S1P3-receptors), and delaying neutrophil apoptosis (neutral sphingomyelinase, S1P1-receptors). They extenuate inflammation by attenuating chemotaxis (S1P) and by stabilizing the endothelial and the epithelial barrier (S1P1-receptor). This chapter discusses the multiple roles and therapeutic options that sphingolipids offer with respect to acute lung injury.
Collapse
Affiliation(s)
- Stefan Uhlig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany.
| | | |
Collapse
|