1
|
Kim HJ, Yang D, Hong JH. Various Cellular Components and Its Signaling Cascades Through the Involvement of Signaling Messengers in Keratinocyte Differentiation. Antioxidants (Basel) 2025; 14:426. [PMID: 40298779 PMCID: PMC12023943 DOI: 10.3390/antiox14040426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/28/2025] [Accepted: 03/30/2025] [Indexed: 04/30/2025] Open
Abstract
Skin is a highly differentiated tissue, in which various signaling molecules play critical roles in the differentiation and proliferation of keratinocytes. Among these, the second messenger calcium and its gradient across skin layers are pivotal in regulating keratinocyte differentiation. Additionally, a diverse array of cellular signaling molecules has been identified as essential for promoting keratinocyte differentiation, thereby maintaining skin integrity and barrier function. The barrier function of the skin provides essential protection against exogenous stimuli and pathogens while maintaining structural stability. The homeostatic processes of skin differentiation are modulated by these second messengers and various signaling molecules. Thus, this review highlights the components associated with keratinocyte differentiation and their biological and pathophysiological roles, as well as redox-sensitive differentiation factors in the modulation of skin homeostasis. This review aims to enhance our understanding of skin physiology and provide insights that may facilitate the development of novel therapeutic strategies for skin diseases.
Collapse
Affiliation(s)
| | - Dongki Yang
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Republic of Korea;
| | - Jeong Hee Hong
- Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, 155 Getbeolro, Yeonsu-gu, Incheon 21999, Republic of Korea;
| |
Collapse
|
2
|
Lee SY, Park JS, Kim D, Jeong W, Hwang C, Kim HO, Park CW, Chung BY. Efficacy of a Multi-lamellar Emulsion Containing a Synthetic Sphingosine Kinase 1 Activator and Pseudoceramide in Patients with Atopic Dermatitis: A Randomized Controlled Trial. Dermatol Ther (Heidelb) 2024; 14:2591-2605. [PMID: 39212849 PMCID: PMC11393266 DOI: 10.1007/s13555-024-01254-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Patients with atopic dermatitis (AD) have impaired barrier function, which decreases skin hydration, weakens their defense against microorganisms, and culminates in increased inflammatory responses. Here, we conducted a clinical trial to evaluate the efficacy of a multi-lamellar emulsion (MLE) containing the pseudoceramide PC-9S and a synthetic sphingosine kinase 1 (SPHK1) activator, Defensamide™, in improving mild-to-moderate atopic dermatitis. METHODS Forty patients aged ≥ 2 years were randomized into a combined-therapy group treated with the MLE containing PC-9S and Defensamide™ plus a topical corticosteroid and a topical-corticosteroid-only group. Assessments based on therapeutic methods included the Eczema Area and Severity Index (EASI), the Investigator Global Assessment (IGA), transepidermal water loss (TEWL), stratum corneum hydration (SCH), skin dryness, a visual analogue scale (VAS) of itchiness, a VAS of sleep disturbance, patient satisfaction, and the Dermatology Life Quality Index (DLQI). RESULTS Thirty-eight patients completed this study. In the combined-therapy group, significant improvements in clinical and instrumental measures such as EASI scores, skin hydration, and skin dryness were noted at 4 weeks compared to baseline, but such improvements were not noted in the topical corticosteroid-only group. Subjective assessments of itching and sleep disturbance and DLQI scores also showed significant improvements in the combined-therapy group. CONCLUSION Combined therapy with the MLE containing Defensamide™ and PC-9S and with topical corticosteroid demonstrated superior clinical outcomes compared with topical corticosteroid monotherapy. Our findings underscore the potential of MLE-containing formulations as effective adjunctive therapies for AD, offering both objective and subjective symptomatic relief and enhancing patients' quality of life.
Collapse
Affiliation(s)
- So Yeon Lee
- Department of Dermatology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, 07441, Republic of Korea
| | - Jin Seo Park
- Department of Dermatology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, 07441, Republic of Korea
| | - Daehwan Kim
- CRID Center, NeoPharm Co., Ltd., Daejeon, Republic of Korea
| | - Wonseok Jeong
- CRID Center, NeoPharm Co., Ltd., Daejeon, Republic of Korea
| | | | - Hye One Kim
- Department of Dermatology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, 07441, Republic of Korea
| | - Chun Wook Park
- Department of Dermatology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, 07441, Republic of Korea
| | - Bo Young Chung
- Department of Dermatology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, 07441, Republic of Korea.
| |
Collapse
|
3
|
Dainese-Marque O, Garcia V, Andrieu-Abadie N, Riond J. Contribution of Keratinocytes in Skin Cancer Initiation and Progression. Int J Mol Sci 2024; 25:8813. [PMID: 39201498 PMCID: PMC11354502 DOI: 10.3390/ijms25168813] [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: 07/19/2024] [Revised: 08/10/2024] [Accepted: 08/11/2024] [Indexed: 09/02/2024] Open
Abstract
Keratinocytes are major cellular components of the skin and are strongly involved in its homeostasis. Oncogenic events, starting mainly from excessive sun exposure, lead to the dysregulation of their proliferation and differentiation programs and promote the initiation and progression of non-melanoma skin cancers (NMSCs). Primary melanomas, which originate from melanocytes, initiate and develop in close interaction with keratinocytes, whose role in melanoma initiation, progression, and immune escape is currently being explored. Recent studies highlighted, in particular, unexpected modes of communication between melanocytic cells and keratinocytes, which may be of interest as sources of new biomarkers in melanomagenesis or potential therapeutic targets. This review aims at reporting the various contributions of keratinocytes in skin basal cell carcinoma (BCC), cutaneous squamous cell carcinoma (cSCC), and melanoma, with a greater focus on the latter in order to highlight some recent breakthrough findings. The readers are referred to recent reviews when contextual information is needed.
Collapse
Affiliation(s)
| | | | - Nathalie Andrieu-Abadie
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, 31037 Toulouse, France
| | - Joëlle Riond
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, 31037 Toulouse, France
| |
Collapse
|
4
|
Zhao F, Zhao J, Wei K, Jiang P, Shi Y, Chang C, Zheng Y, Shan Y, Li Y, He B, Zhou M, Liu J, Li L, Guo S, He D. Targeted siRNA Therapy for Psoriasis: Translating Preclinical Potential into Clinical Treatments. Immunotargets Ther 2024; 13:259-271. [PMID: 38770264 PMCID: PMC11104385 DOI: 10.2147/itt.s458800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by the excessive proliferation of keratinocytes and heightened immune activation. Targeting pathogenic genes through small interfering RNA (siRNA) therapy represents a promising strategy for the treatment of psoriasis. This mini-review provides a comprehensive summary of siRNA research targeting the pathogenesis of psoriasis, covering aspects such as keratinocyte function, inflammatory cell roles, preclinical animal studies, and siRNA delivery mechanisms. It details recent advancements in RNA interference that modulate key factors including keratinocyte proliferation (Fibroblast Growth Factor Receptor 2, FGFR2), apoptosis (Interferon Alpha Inducible Protein 6, G1P3), differentiation (Grainyhead Like Transcription Factor 2, GRHL2), and angiogenesis (Vascular Endothelial Growth Factor, VEGF); immune cell infiltration and inflammation (Tumor Necrosis Factor-Alpha, TNF-α; Interleukin-17, IL-17); and signaling pathways (JAK-STAT, Nuclear Factor Kappa B, NF-κB) that govern immunopathology. Despite significant advances in siRNA-targeted treatments for psoriasis, several challenges persist. Continued scientific developments promise the creation of more effective and safer siRNA medications, potentially enhancing the quality of life for psoriasis patients and revolutionizing treatments for other diseases. This article focuses on the most recent research advancements in targeting the pathogenesis of psoriasis with siRNA and explores its future therapeutic prospects.
Collapse
Affiliation(s)
- Fuyu Zhao
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jianan Zhao
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Kai Wei
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Ping Jiang
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yiming Shi
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Cen Chang
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yixin Zheng
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yu Shan
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yunshen Li
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Bingheng He
- Department of Rehabilitation, Tongren Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Mi Zhou
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jia Liu
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Li Li
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Shicheng Guo
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Dongyi He
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
5
|
Jin L, Zhu J, Yao L, Shen G, Xue BX, Tao W. Targeting SphK1/2 by SKI-178 inhibits prostate cancer cell growth. Cell Death Dis 2023; 14:537. [PMID: 37604912 PMCID: PMC10442381 DOI: 10.1038/s41419-023-06023-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 08/23/2023]
Abstract
Sphingosine kinases (SphK), including SphK1 and SphK2, are important enzymes promoting progression of prostate cancer. SKI-178 is a novel and highly potent SphK1/2 dual inhibitor. We here tested the potential anti-prostate cancer cell activity of SKI-178. Bioinformatics analyses and results from local tissues demonstrated that that both SphK1 and SphK2 are upregulated in human prostate cancer tissues. Ectopic overexpression of SphK1 and SphK2, by lentiviral constructs, promoted primary prostate cancer cell proliferation and migration. In primary human prostate cancer cells and immortalized cell lines, SKI-178 potently inhibited cell viability, proliferation, cell cycle progression and cell migration, causing robust cell death and apoptosis. SKI-178 impaired mitochondrial functions, causing mitochondrial depolarization, reactive oxygen species production and ATP depletion.SKI-178 potently inhibited SphK activity and induced ceramide production, without affecting SphK1/2 expression in prostate cancer cells. Further, SKI-178 inhibited Akt-mTOR activation and induced JNK activation in prostate cancer cells. Contrarily, a constitutively-active Akt1 construct or the pharmacological JNK inhibitors attenuated SKI-178-induced cytotoxicity in prostate cancer cells. In vivo, daily intraperitoneal injection of a single dose of SKI-178 potently inhibited PC-3 xenograft growth in nude mice. SphK inhibition, ceramide production, ATP depletion and lipid peroxidation as well as Akt-mTOR inactivation and JNK activation were detected in PC-3 xenograft tissues with SKI-178 administration. Together, targeting SphK1/2 by SKI-178 potently inhibited prostate cancer cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Lu Jin
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Zhu
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Linya Yao
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine Affiliated to Yangzhou University, Kunshan, China
| | - Gang Shen
- Department of Urology, DUSHU Lake Hospital Affiliated to Soochow University, Suzhou, China.
| | - Bo-Xin Xue
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Wei Tao
- Department of Urology, the Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
6
|
Smith CJ, Williams JL, Hall C, Casas J, Caley MP, O'Toole EA, Prasad R, Metherell LA. Ichthyosis linked to sphingosine 1-phosphate lyase insufficiency is due to aberrant sphingolipid and calcium regulation. J Lipid Res 2023; 64:100351. [PMID: 36868360 PMCID: PMC10123262 DOI: 10.1016/j.jlr.2023.100351] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 03/05/2023] Open
Abstract
Sphingosine 1-phosphate lyase (SGPL1) insufficiency (SPLIS) is a syndrome which presents with adrenal insufficiency, steroid-resistant nephrotic syndrome, hypothyroidism, neurological disease, and ichthyosis. Where a skin phenotype is reported, 94% had abnormalities such as ichthyosis, acanthosis, and hyperpigmentation. To elucidate the disease mechanism and the role SGPL1 plays in the skin barrier we established clustered regularly interspaced short palindromic repeats-Cas9 SGPL1 KO and a lentiviral-induced SGPL1 overexpression (OE) in telomerase reverse-transcriptase immortalised human keratinocytes (N/TERT-1) and thereafter organotypic skin equivalents. Loss of SGPL1 caused an accumulation of S1P, sphingosine, and ceramides, while its overexpression caused a reduction of these species. RNAseq analysis showed perturbations in sphingolipid pathway genes, particularly in SGPL1_KO, and our gene set enrichment analysis revealed polar opposite differential gene expression between SGPL1_KO and _OE in keratinocyte differentiation and Ca2+ signaling genesets. SGPL1_KO upregulated differentiation markers, while SGPL1_OE upregulated basal and proliferative markers. The advanced differentiation of SGPL1_KO was confirmed by 3D organotypic models that also presented with a thickened and retained stratum corneum and a breakdown of E-cadherin junctions. We conclude that SPLIS associated ichthyosis is a multifaceted disease caused possibly by sphingolipid imbalance and excessive S1P signaling, leading to increased differentiation and an imbalance of the lipid lamellae throughout the epidermis.
Collapse
Affiliation(s)
- Christopher J Smith
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
| | - Jack L Williams
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Charlotte Hall
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain; Biomedical Research Centre (CIBEREHD), ISCIII, Madrid, Spain
| | - Matthew P Caley
- Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Edel A O'Toole
- Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Rathi Prasad
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Louise A Metherell
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
7
|
Kleuser B, Bäumer W. Sphingosine 1-Phosphate as Essential Signaling Molecule in Inflammatory Skin Diseases. Int J Mol Sci 2023; 24:ijms24021456. [PMID: 36674974 PMCID: PMC9863039 DOI: 10.3390/ijms24021456] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Sphingolipids are crucial molecules of the mammalian epidermis. The formation of skin-specific ceramides contributes to the formation of lipid lamellae, which are important for the protection of the epidermis from excessive water loss and protect the skin from the invasion of pathogens and the penetration of xenobiotics. In addition to being structural constituents of the epidermal layer, sphingolipids are also key signaling molecules that participate in the regulation of epidermal cells and the immune cells of the skin. While the importance of ceramides with regard to the proliferation and differentiation of skin cells has been known for a long time, it has emerged in recent years that the sphingolipid sphingosine 1-phosphate (S1P) is also involved in processes such as the proliferation and differentiation of keratinocytes. In addition, the immunomodulatory role of this sphingolipid species is becoming increasingly apparent. This is significant as S1P mediates a variety of its actions via G-protein coupled receptors. It is, therefore, not surprising that dysregulation in the signaling pathways of S1P is involved in the pathophysiological conditions of skin diseases. In the present review, the importance of S1P in skin cells, as well as the immune cells of the skin, is elaborated. In particular, the role of the molecule in inflammatory skin diseases will be discussed. This is important because interfering with S1P signaling pathways may represent an innovative option for the treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Burkhard Kleuser
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise Str. 2+4, 14195 Berlin, Germany
- Correspondence: (B.K.); (W.B.)
| | - Wolfgang Bäumer
- Department of Veterinary Medicine, Institute of Pharmacology and Toxicology, Freie Universität Berlin, Koserstr. 20, 14195 Berlin, Germany
- Correspondence: (B.K.); (W.B.)
| |
Collapse
|
8
|
Matwiejuk M, Mysliwiec H, Chabowski A, Flisiak I. The Role of Sphingolipids in the Pathogenesis of Psoriasis. Metabolites 2022; 12:1171. [PMID: 36557209 PMCID: PMC9785224 DOI: 10.3390/metabo12121171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Psoriasis is a complex, chronic, immunologically mediated disease which involves skin and joints. Psoriasis is commonly connected with numerous other diseases such as liver diseases, metabolic syndrome, impaired glucose tolerance, diabetes mellitus, atherosclerosis, hypertension, and ischemic heart disease. Interestingly, comorbidities of psoriasis are an attention-grabbing issue. Additionally, it can cause impairment of quality of life and may be associated with depressive disorders. Altered levels of ceramides in psoriatic skin may lead to anti-apoptotic and pro-proliferative states, consequently leading to an over-proliferation of keratinocytes and the development of skin lesions. The pathophysiology of psoriasis and its comorbidities is not fully understood yet. Sphingolipids (including ceramides) and their disturbed metabolism may be the link between psoriasis and its comorbidities. Overall, the goal of this review was to discuss the role of sphingolipid disturbances in psoriasis and its comorbidities. We searched the PubMed database for relevant articles published before the beginning of May 2022. The systematic review included 65 eligible original articles.
Collapse
Affiliation(s)
- Mateusz Matwiejuk
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland
| | - Hanna Mysliwiec
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, 15-222 Bialystok, Poland
| | - Iwona Flisiak
- Department of Dermatology and Venereology, Medical University of Bialystok, 15-540 Bialystok, Poland
| |
Collapse
|
9
|
Targeting sphingosine kinase 1/2 by a novel dual inhibitor SKI-349 suppresses non-small cell lung cancer cell growth. Cell Death Dis 2022; 13:602. [PMID: 35831279 PMCID: PMC9279331 DOI: 10.1038/s41419-022-05049-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/16/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Sphingosine kinase 1 (SphK1) and sphingosine kinase (SphK2) are both important therapeutic targets of non-small cell lung cancer (NSCLC). SKI-349 is a novel, highly efficient and small molecular SphK1/2 dual inhibitor. Here in primary human NSCLC cells and immortalized cell lines, SKI-349 potently inhibited cell proliferation, cell cycle progression, migration and viability. The dual inhibitor induced mitochondrial depolarization and apoptosis activation in NSCLC cells, but it was non-cytotoxic to human lung epithelial cells. SKI-349 inhibited SphK activity and induced ceramide accumulation in primary NSCLC cells, without affecting SphK1/2 expression. SKI-349-induced NSCLC cell death was attenuated by sphingosine-1-phosphate and by the SphK activator K6PC-5, but was potentiated by the short-chain ceramide C6. Moreover, SKI-349 induced Akt-mTOR inactivation, JNK activation, and oxidative injury in primary NSCLC cells. In addition, SKI-349 decreased bromodomain-containing protein 4 (BRD4) expression and downregulated BRD4-dependent genes (Myc, cyclin D1 and Klf4) in primary NSCLC cells. At last, SKI-349 (10 mg/kg) administration inhibited NSCLC xenograft growth in nude mice. Akt-mTOR inhibition, JNK activation, oxidative injury and BRD4 downregulation were detected in SKI-349-treated NSCLC xenograft tissues. Taken together, targeting SphK1/2 by SKI-349 potently inhibits NSCLC cell growth in vitro and in vivo.
Collapse
|
10
|
Sun X, Shan HJ, Yin G, Zhang XY, Huang YM, Li HJ. The anti-osteosarcoma cell activity by the sphingosine kinase 1 inhibitor SKI-V. Cell Death Dis 2022; 8:48. [PMID: 35115496 PMCID: PMC8814198 DOI: 10.1038/s41420-022-00838-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/03/2022] [Accepted: 01/14/2022] [Indexed: 01/04/2023]
Abstract
Sphingosine kinase 1 (SphK1) expression and activity are elevated in human osteosarcoma (OS) and is a promising target of therapy. SKI-V is a non-competitive and highly-efficient non-lipid SphK1 inhibitor. The potential anti-OS cell activity by the SphK1 inhibitor was studied here. In primary OS cells and immortalized cell lines, SKI-V robustly suppressed cell survival, growth and proliferation as well as cell mobility, and inducing profound OS cell death and apoptosis. The SphK1 inhibitor was however non-cytotoxic nor pro-apoptotic in human osteoblasts. SKI-V robustly inhibited SphK1 activation and induced accumulation of ceramides, without affecting SphK1 expression in primary OS cells. The SphK1 activator K6PC-5 or sphingosine-1-phosphate partially inhibited SKI-V-induced OS cell death. We showed that SKI-V concurrently blocked Akt-mTOR activation in primary OS cells. A constitutively-active Akt1 (ca-Akt1, S473D) construct restored Akt-mTOR activation and mitigated SKI-V-mediated cytotoxicity in primary OS cells. In vivo, daily injection of SKI-V potently suppressed OS xenograft tumor growth in nude mice. In SKI-V-administrated OS xenograft tissues, SphK1 inhibition, ceramide increase and Akt-mTOR inhibition were detected. Together, SKI-V exerts significant anti-OS activity by inhibiting SphK1 and Akt-mTOR cascades in OS cells.
Collapse
Affiliation(s)
- Xu Sun
- Department of Hand and Foot Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, Taizhou, China
| | - Hua-Jian Shan
- Department of Orthopaedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Yin
- Department of Orthopaedics, Wujin Hospital Affiliated to Jiangsu University, Changzhou, China
| | - Xiang-Yang Zhang
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Min Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Hai-Jun Li
- Department of Hand and Foot Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, Taizhou, China.
| |
Collapse
|
11
|
Vietri Rudan M, Watt FM. Mammalian Epidermis: A Compendium of Lipid Functionality. Front Physiol 2022; 12:804824. [PMID: 35095565 PMCID: PMC8791442 DOI: 10.3389/fphys.2021.804824] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022] Open
Abstract
Mammalian epidermis is a striking example of the role of lipids in tissue biology. In this stratified epithelium, highly specialized structures are formed that leverage the hydrophobic properties of lipids to form an impermeable barrier and protect the humid internal environment of the body from the dry outside. This is achieved through tightly regulated lipid synthesis that generates the molecular species unique to the tissue. Beyond their fundamental structural role, lipids are involved in the active protection of the body from external insults. Lipid species present on the surface of the body possess antimicrobial activity and directly contribute to shaping the commensal microbiota. Lipids belonging to a variety of classes are also involved in the signaling events that modulate the immune responses to environmental stress as well as differentiation of the epidermal keratinocytes themselves. Recently, high-resolution methods are beginning to provide evidence for the involvement of newly identified specific lipid molecules in the regulation of epidermal homeostasis. In this review we give an overview of the wide range of biological functions of mammalian epidermal lipids.
Collapse
|
12
|
Imre G, Krähling V, Eichler M, Trautmann S, Ferreirós N, Aman MJ, Kashanchi F, Rajalingam K, Pöhlmann S, Becker S, Meyer Zu Heringdorf D, Pfeilschifter J. The sphingosine kinase 1 activator, K6PC-5, attenuates Ebola virus infection. iScience 2021; 24:102266. [PMID: 33817572 PMCID: PMC8005759 DOI: 10.1016/j.isci.2021.102266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 02/08/2021] [Accepted: 03/01/2021] [Indexed: 12/18/2022] Open
Abstract
Ebola virus (EBOV) is responsible for outbreaks with case fatality rates of up to 90% and for an epidemic in West Africa with more than ten thousand deaths. EBOV glycoprotein (EBOV-GP) is the only viral surface protein and is responsible for viral entry into cells. Here, by employing pseudotyped EBOV-GP viral particles, we uncover a critical role for sphingolipids in inhibiting viral entry. Sphingosine kinase 1 (SphK1) catalyzes the phosphorylation of sphingosine to sphingosine 1-phosphate (S1P). The administration of the SphK1 activator, K6PC-5, or S1P, or the overexpression of SphK1 consistently exhibited striking inhibitory effects in EBOV-GP-driven entry in diverse cell lines. Finally, K6PC-5 markedly reduced the EBOV titer in infected cells and the de novo production of viral proteins. These data present K6PC-5 as an efficient tool to inhibit EBOV infection in endothelial cells and suggest further studies to evaluate its systemic effects. K6PC-5, a sphingosine kinase 1 activator, inhibits Ebola virus infection Sphingosine 1-phosphate, the product of SphK1, attenuates the viral entry Inhibiton/activation of S1P receptors has no influence on Ebola virus entry These data support the endogen effect of S1P in Ebola virus infection
Collapse
Affiliation(s)
- Gergely Imre
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Verena Krähling
- Institute of Virology, Philipps University Marburg, Marburg, Germany.,German Center for Infection Research (DZIF), partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Madeleine Eichler
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Sandra Trautmann
- Institute of Clinical Pharmacology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Nerea Ferreirós
- Institute of Clinical Pharmacology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - M Javad Aman
- Integrated BioTherapeutics, Inc., Gaithersburg, MD 20850, USA
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, George Mason University Manassas, VA 20110, USA
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, 37077 Göttingen, Germany.,Faculty of Biology and Psychology, University Göttingen, 37077 Göttingen, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, Marburg, Germany.,German Center for Infection Research (DZIF), partner site Gießen-Marburg-Langen, Marburg, Germany
| | - Dagmar Meyer Zu Heringdorf
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Josef Pfeilschifter
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| |
Collapse
|
13
|
Xie T, Chen C, Peng Z, Brown BC, Reisz JA, Xu P, Zhou Z, Song A, Zhang Y, Bogdanov MV, Kellems RE, D'Alessandro A, Zhang W, Xia Y. Erythrocyte Metabolic Reprogramming by Sphingosine 1-Phosphate in Chronic Kidney Disease and Therapies. Circ Res 2020; 127:360-375. [PMID: 32284030 DOI: 10.1161/circresaha.119.316298] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
RATIONALE Hypoxia promotes renal damage and progression of chronic kidney disease (CKD). The erythrocyte is the only cell type for oxygen (O2) delivery. Sphingosine 1-phosphate (S1P)-a highly enriched biolipid in erythrocytes-is recently reported to be induced under high altitude in normal humans to enhance O2 delivery. However, nothing is known about erythrocyte S1P in CKD. OBJECTIVE To investigate the function and metabolic basis of erythrocyte S1P in CKD with a goal to explore potential therapeutics. METHODS AND RESULTS Using erythrocyte-specific SphK1 (sphingosine kinase 1; the only enzyme to produce S1P in erythrocytes) knockout mice (eSphK1-/-) in an experimental model of hypertensive CKD with Ang II (angiotensin II) infusion, we found severe renal hypoxia, hypertension, proteinuria, and fibrosis in Ang II-infused eSphk1-/- mice compared with controls. Untargeted metabolomics profiling and in vivo U-13C6 isotopically labeled glucose flux analysis revealed that SphK1 is required for channeling glucose metabolism toward glycolysis versus pentose phosphate pathway, resulting in enhanced erythroid-specific Rapoport-Luebering shunt in Ang II-infused mice. Mechanistically, increased erythrocyte S1P functioning intracellularly activates AMPK (AMP-activated protein kinase) 1α and BPGM (bisphosphoglycerate mutase) by reducing ceramide/S1P ratio and inhibiting PP2A (protein phosphatase 2A), leading to increased 2,3-bisphosphoglycerate (an erythrocyte-specific metabolite negatively regulating Hb [hemoglobin]-O2-binding affinity) production and thus more O2 delivery to counteract kidney hypoxia and progression to CKD. Preclinical studies revealed that an AMPK agonist or a PP2A inhibitor rescued the severe CKD phenotype in Ang II-infused eSphK1-/- mice and prevented development of CKD in the control mice by inducing 2,3-bisphosphoglycerate production and thus enhancing renal oxygenation. Translational research validated mouse findings in erythrocytes of hypertensive CKD patients and cultured human erythrocytes. CONCLUSIONS Our study elucidates the beneficial role of eSphk1-S1P in hypertensive CKD by channeling glucose metabolism toward Rapoport-Luebering shunt and inducing 2,3-bisphosphoglycerate production and O2 delivery via a PP2A-AMPK1α signaling pathway. These findings reveal the metabolic and molecular basis of erythrocyte S1P in CKD and new therapeutic avenues.
Collapse
Affiliation(s)
- Tingting Xie
- From the Rheumatology and Immunology (T.X.), Xiangya Hospital, Central South University, Changsha, Hunan, China.,Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Changhan Chen
- Otolaryngology Head and Neck Surgery (C.C.), Xiangya Hospital, Central South University, Changsha, Hunan, China.,Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Zhangzhe Peng
- Nephrology (Z.P.), Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Benjamin C Brown
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora (B.C.B., J.A.R., A.D.)
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora (B.C.B., J.A.R., A.D.)
| | - Ping Xu
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Zhen Zhou
- Division of Medical Genetics, Department of Internal Medicine (Z.Z.), University of Texas McGovern Medical School at Houston
| | - Anren Song
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Yujin Zhang
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Mikhail V Bogdanov
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Rodney E Kellems
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston.,MDAnderson-UTHealth Graduate School of Biomedical Science, Houston, TX (R.E.K., Y.X.)
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora (B.C.B., J.A.R., A.D.)
| | - Weiru Zhang
- General Medicine (W.Z.), Xiangya Hospital, Central South University, Changsha, Hunan, China.,Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston
| | - Yang Xia
- Biochemistry and Molecular Biology (T.X., C.C., P.X., A.S., Y.Z., M.V.B., R.E.K., W.Z., Y.X.), University of Texas McGovern Medical School at Houston.,MDAnderson-UTHealth Graduate School of Biomedical Science, Houston, TX (R.E.K., Y.X.)
| |
Collapse
|
14
|
Di Pardo A, Pepe G, Capocci L, Marracino F, Amico E, Del Vecchio L, Giova S, Jeong SK, Park BM, Park BD, Maglione V. Treatment with K6PC-5, a selective stimulator of SPHK1, ameliorates intestinal homeostasis in an animal model of Huntington's disease. Neurobiol Dis 2020; 143:105009. [PMID: 32634578 DOI: 10.1016/j.nbd.2020.105009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/10/2020] [Accepted: 06/29/2020] [Indexed: 02/08/2023] Open
Abstract
Emerging evidence indicates that Huntington's disease (HD) may be described as multi-organ pathology. In this context, we and others have contributed to demonstrate that the disease is characterized by an impairment of the homeostasis of gastro-intestinal (GI) tract. Sphingolipids represent a class of molecules involved in the regulation and maintenance of different tissues and organs including GI system. In this study, we investigated whether the alteration of Sphingosine-1-phosphate (S1P) metabolism, previously described in human HD brains and animal models, is also detectable peripherally in R6/2 HD mice. Our findings indicate, for the first time, that sphingolipid metabolism is perturbed early in the disease in the intestinal tract of HD mice and, its modulation by K6PC-5, a selective activator of S1P synthesis, preserved intestinal integrity and homeostasis. These results further support the evidence that modulation of sphingolipid pathways may represent a potential therapeutic option in HD and suggest that it has also the potential to counteract the peripheral disturbances which may usually complicate the management of the disease and affect patient's quality of life.
Collapse
Affiliation(s)
| | - G Pepe
- IRCCS Neuromed, Pozzilli, Italy
| | | | | | - E Amico
- IRCCS Neuromed, Pozzilli, Italy
| | - L Del Vecchio
- IRCCS Neuromed, Pozzilli, Italy; Unità complessa di radiodiagnostica (U.O.C.) POS, University of Foggia, Foggia, Italy
| | - S Giova
- IRCCS Neuromed, Pozzilli, Italy
| | - S K Jeong
- Department of Cosmetic Science, Seowon University, Cheongju, Republic of Korea
| | - B M Park
- NeoPharm USA Inc., Engelwood Cliffs, NJ, USA
| | - B D Park
- Dr. Raymond Laboratories, Inc, Englewood cliffs, NJ, USA
| | | |
Collapse
|
15
|
Jeon S, Song J, Lee D, Kim GT, Park SH, Shin DY, Shin KO, Park K, Shim SM, Park TS. Inhibition of sphingosine 1-phosphate lyase activates human keratinocyte differentiation and attenuates psoriasis in mice. J Lipid Res 2020; 61:20-32. [PMID: 31690639 PMCID: PMC6939600 DOI: 10.1194/jlr.ra119000254] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/29/2019] [Indexed: 11/20/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) lyase is an intracellular enzyme that catalyzes the irreversible degradation of S1P and has been suggested as a therapeutic target for the treatment of psoriasis vulgaris. Because S1P induces differentiation of keratinocytes, we examined whether modulation of S1P lyase and altered intracellular S1P levels regulate proliferation and differentiation of human neonatal epidermal keratinocyte (HEKn) cells. To identify the physiological functions of S1P lyase in skin, we inhibited S1P lyase in HEKn cells with an S1P lyase-specific inhibitor (SLI) and with S1P lyase 1 (SGPL1)-specific siRNA (siSGPL1). In HEKn cells, pharmacological treatment with the SLI caused G1 arrest by upregulation of p21 and p27 and induced keratin 1, an early differentiation marker. Similarly, genetic suppression by siSGPL1 arrested the cell cycle at the G1 phase and activated differentiation. In addition, enzyme suppression by siSGPL1 upregulated keratin 1 and differentiation markers including involucrin and loricrin. When hyperproliferation of HEKn cells was induced by interleukin (IL)-17 and IL-22, pharmacologic inhibition of S1P lyase by SLI decreased proliferation and activated differentiation of HEKn cells simultaneously. In addition, SLI administration ameliorated imiquimod-induced psoriatic symptoms including erythema, scaling, and epidermal thickness in vivo. We thus demonstrated that S1P lyase inhibition reduces cell proliferation and induces keratinocyte differentiation, and that inhibition may attenuate psoriasiform changes. Collectively, these findings suggest that S1P lyase is a modulating factor for proliferation and differentiation, and support its potential as a therapeutic target for psoriasis in human keratinocytes.
Collapse
Affiliation(s)
- Suwon Jeon
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Jaehwi Song
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Dongyup Lee
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Goon-Tae Kim
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Si-Hyun Park
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Dong-Yoon Shin
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Kyong-Oh Shin
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kyungho Park
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Soon-Mi Shim
- Department of Food Science and Technology, Sejong University, Seoul 05006, Republic of Korea.
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea.
| |
Collapse
|
16
|
Defective Sphingosine-1-phosphate metabolism is a druggable target in Huntington's disease. Sci Rep 2017; 7:5280. [PMID: 28706199 PMCID: PMC5509685 DOI: 10.1038/s41598-017-05709-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/01/2017] [Indexed: 12/22/2022] Open
Abstract
Huntington’s disease is characterized by a complex and heterogeneous pathogenic profile. Studies have shown that disturbance in lipid homeostasis may represent a critical determinant in the progression of several neurodegenerative disorders. The recognition of perturbed lipid metabolism is only recently becoming evident in HD. In order to provide more insight into the nature of such a perturbation and into the effect its modulation may have in HD pathology, we investigated the metabolism of Sphingosine-1-phosphate (S1P), one of the most important bioactive lipids, in both animal models and patient samples. Here, we demonstrated that S1P metabolism is significantly disrupted in HD even at early stage of the disease and importantly, we revealed that such a dysfunction represents a common denominator among multiple disease models ranging from cells to humans through mouse models. Interestingly, the in vitro anti-apoptotic and the pro-survival actions seen after modulation of S1P-metabolizing enzymes allows this axis to emerge as a new druggable target and unfolds its promising therapeutic potential for the development of more effective and targeted interventions against this incurable condition.
Collapse
|
17
|
Zhang S, Deng Z, Yao C, Huang P, Zhang Y, Cao S, Li X. AT7867 Inhibits Human Colorectal Cancer Cells via AKT-Dependent and AKT-Independent Mechanisms. PLoS One 2017; 12:e0169585. [PMID: 28081222 PMCID: PMC5231330 DOI: 10.1371/journal.pone.0169585] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/19/2016] [Indexed: 11/18/2022] Open
Abstract
AKT is often hyper-activated in human colorectal cancers (CRC). This current study evaluated the potential anti-CRC activity by AT7867, a novel AKT and p70S6K1 (S6K1) dual inhibitor. We showed that AT7867 inhibited survival and proliferation of established (HT-29, HCT116 and DLD-1 lines) and primary human CRC cells. Meanwhile, it provoked caspase-dependent apoptosis in the CRC cells. Molecularly, AT7867 blocked AKT-S6K1 activation in CRC cells. Restoring AKT-S6K1 activation, via expression of a constitutively-active AKT1 ("ca-AKT1"), only partially attenuated AT7867-induced HT-29 cell death. Further studies demonstrated that AT7867 inhibited sphingosine kinase 1 (SphK1) activity to promote pro-apoptotic ceramide production in HT-29 cells. Such effects by AT7867 were independent of AKT inhibition. AT7867-indued ceramide production and subsequent HT-29 cell apoptosis were attenuated by co-treatment of sphingosine-1-phosphate (S1P), but were potentiated with the glucosylceramide synthase (GCS) inhibitor PDMP. In vivo, intraperitoneal injection of AT7867 inhibited HT-29 xenograft tumor growth in nude mice. AKT activation was also inhibited in AT7867-treated HT-29 tumors. Together, the preclinical results suggest that AT7867 inhibits CRC cells via AKT-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Shihu Zhang
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhengming Deng
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen Yao
- Orthopedic Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Ping Huang
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Zhang
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shibing Cao
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiangcheng Li
- Department of General Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Ebenezer DL, Fu P, Suryadevara V, Zhao Y, Natarajan V. Epigenetic regulation of pro-inflammatory cytokine secretion by sphingosine 1-phosphate (S1P) in acute lung injury: Role of S1P lyase. Adv Biol Regul 2017; 63:156-166. [PMID: 27720306 PMCID: PMC5292070 DOI: 10.1016/j.jbior.2016.09.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 09/20/2016] [Accepted: 09/25/2016] [Indexed: 11/25/2022]
Abstract
Cellular level of sphingosine-1-phosphate (S1P), the simplest bioactive sphingolipid, is tightly regulated by its synthesis catalyzed by sphingosine kinases (SphKs) 1 & 2 and degradation mediated by S1P phosphatases, lipid phosphate phosphatases, and S1P lyase. The pleotropic actions of S1P are attributed to its unique inside-out (extracellular) signaling via G-protein-coupled S1P1-5 receptors, and intracellular receptor independent signaling. Additionally, S1P generated in the nucleus by nuclear SphK2 modulates HDAC1/2 activity, regulates histone acetylation, and transcription of pro-inflammatory genes. Here, we present data on the role of S1P lyase mediated S1P signaling in regulating LPS-induced inflammation in lung endothelium. Blocking S1P lyase expression or activity attenuated LPS-induced histone acetylation and secretion of pro-inflammatory cytokines. Degradation of S1P by S1P lyase generates Δ2-hexadecenal and ethanolamine phosphate and the long-chain fatty aldehyde produced in the cytoplasmic compartment of the endothelial cell seems to modulate histone acetylation pattern, which is different from the nuclear SphK2/S1P signaling and inhibition of HDAC1/2. These in vitro studies suggest that S1P derived long-chain fatty aldehyde may be an epigenetic regulator of pro-inflammatory genes in sepsis-induced lung inflammation. Trapping fatty aldehydes and other short chain aldehydes such as 4-hydroxynonenal derived from S1P degradation and lipid peroxidation, respectively by cell permeable agents such as phloretin or other aldehyde trapping agents may be useful in treating sepsis-induced lung inflammation via modulation of histone acetylation. .
Collapse
Affiliation(s)
- David L Ebenezer
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Panfeng Fu
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Vidyani Suryadevara
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Yutong Zhao
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA; Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
19
|
Xu L, Zhang Y, Gao M, Wang G, Fu Y. Concurrent targeting Akt and sphingosine kinase 1 by A-674563 in acute myeloid leukemia cells. Biochem Biophys Res Commun 2016; 472:662-8. [PMID: 26920060 DOI: 10.1016/j.bbrc.2016.02.094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/22/2016] [Indexed: 12/13/2022]
Abstract
Akt signaling plays a pivotal role in acute myeloid leukemia (AML) development and progression. In the present study, we evaluated the potential anti-AML activity by a novel Akt kinase inhibitor A-674563. Our results showed that A-674563 dose-dependently inhibited survival and proliferation of U937 AML cells and six lines of human AML progenitor cells, yet sparing human peripheral blood mononuclear leukocytes (PBMCs). A-674563 activated caspase-3/9 and apoptosis in the AML cells. Reversely, the pan-caspase inhibitor z-VAD-CHO dramatically alleviated A-674563-induced AML cell apoptosis and cytotoxicity. For the molecular study, we showed that A-674563 blocked Akt activation in U937 cells and human AML progenitor cells. Further, A-674563 decreased sphingosine kinase 1 (SphK1) activity in above AML cells to deplete pro-survival sphingosine-1-phosphate (S1P) and boost pro-apoptotic ceramide production. Such an effect on SphK1 signaling by A-674563 appeared independent of Akt blockage. Significantly, K6PC-5, a novel SphK1 activator, or supplement with S1P attenuated A-674563-induced ceramide production, and subsequent U937 cell death and apoptosis. Importantly, intraperitoneal injection of A-674563 at well-tolerated doses suppressed U937 leukemic xenograft tumor growth in nude mice, whiling significantly improving the animal survival. The results of the current study demonstrate that A-674563 exerts potent anti-leukemic activity in vitro and in vivo, possibly via concurrent targeting Akt and SphK1 signalings.
Collapse
Affiliation(s)
- Lin Xu
- Xiangya Hospital, Central South University, Changsha, China; Shaoyang Central Hospital, Hunan Province, China
| | - Yanan Zhang
- The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Meng Gao
- The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Guangping Wang
- Xiangya Hospital, Central South University, Changsha, China.
| | - Yunfeng Fu
- The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
20
|
Guo S, Xie Y, Fan JB, Ji F, Wang S, Fei H. α-Melanocyte stimulating hormone attenuates dexamethasone-induced osteoblast damages through activating melanocortin receptor 4-SphK1 signaling. Biochem Biophys Res Commun 2015; 469:281-7. [PMID: 26631960 DOI: 10.1016/j.bbrc.2015.11.104] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 11/23/2015] [Indexed: 11/25/2022]
Abstract
Long-term glucocorticoid (GC) usage may cause non-traumatic femoral head osteonecrosis. Dexamethasone (Dex) is shown to exert potent cytotoxic effect to osteoblasts. Here, we investigated the potential activity of α-melanocyte stimulating hormone (α-MSH) against the process. Our data revealed that pretreatment of α-MSH significantly inhibited Dex-induced apoptosis and necrosis in both osteoblastic-like MC3T3-E1 cells and primary murine osteoblasts. Melanocortin receptor 4 (MC4R) acts as the receptor of α-MSH in mediating its actions in osteoblasts. The MC4R antagonist SHU9119, or shRNA-mediated knockdown of MC4R, almost abolished α-MSH-induced activation of downstream signalings (Akt and Erk1/2) and its pro-survival effect in osteoblasts. Further studies showed that α-MSH activated MC4R downstream sphingosine kinase 1 (SphK1) and increased cellular sphingosine-1-phosphate (S1P) content in MC3T3-E1 cells and primary murine osteoblasts, which were blocked by SHU9119 or MC4R shRNAs. SphK1 inhibition by the its inhibitor N,N-dimethylsphingosine (DMS), or SphK1 knockdown by targeted-shRNAs, largely attenuated α-MSH-mediated osteoblast protection against Dex. Together, these results suggest that α-MSH alleviates Dex-induced damages to cultured osteoblasts through activating MC4R-SphK1 signaling.
Collapse
Affiliation(s)
- Shiguang Guo
- Department of Intensive Care Unit, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, PR China
| | - Yue Xie
- Department of Orthopedics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, PR China
| | - Jian-bo Fan
- The Department of Orthopaedics, The Second Affiliated Hospital of Nantong University, Nantong, PR China
| | - Feng Ji
- Department of Orthopedics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, PR China.
| | - Shouguo Wang
- Department of Orthopedics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, PR China
| | - Haodong Fei
- Department of Orthopedics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, PR China
| |
Collapse
|
21
|
Shao JJ, Peng Y, Wang LM, Wang JK, Chen X. Activation of SphK1 by K6PC-5 Inhibits Oxygen-Glucose Deprivation/Reoxygenation-Induced Myocardial Cell Death. DNA Cell Biol 2015; 34:669-76. [PMID: 26308910 DOI: 10.1089/dna.2015.2959] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
In the current study, we evaluated the potential effect of a novel sphingosine kinase 1 (SphK1) activator, K6PC-5, on oxygen-glucose deprivation (OGD)/reoxygenation-induced damages to myocardial cells. We demonstrated that K6PC-5 increased intracellular sphingosine-1-phosphate (S1P) content and remarkably inhibited OGD/reoxygenation-induced death of myocardial cells (H9c2/HL-1 lines and primary murine myocardiocytes). SphK1 inhibitors, B-5354c and SKI-II, or SphK1-siRNA knockdown not only aggregated OGD/reoxygenation-induced cytotoxicity but also nullified the cytoprotection by K6PC-5. On the other hand, overexpression of SphK1 alleviated H9c2 cell death by OGD/reoxygenation, and K6PC-5-mediated cytoprotection was also enhanced in SphK1 overexpressed cells. Molecularly, OGD/reoxygenation activated the mitochondrial death pathway, evidenced by reactive oxygen species (ROS) production, mitochondrial membrane potential reduction, and p53-cyclophilin D (Cyp-D) association, which were all alleviated by K6PC-5 or overexpression of SphK1, but exacerbated by SphK1 knockdown. Furthermore, OGD/reoxygenation induced prodeath ceramide production in myocardial cells, which was largely suppressed by K6PC-5. In the meantime, adding a cell-permeable short-chain ceramide (C6) mimicked OGD/reoxygenation actions and induced ROS production and the mitochondrial death pathway in myocardial cells. Together, we conclude that K6PC-5 inhibits OGD/reoxygenation-induced myocardial cell death probably through activating SphK1. The results of the study indicate a potential benefit of K6PC-5 on ischemic heart disease.
Collapse
Affiliation(s)
- Jun-jie Shao
- 1 Department of Cardiothoracic Surgery, Nanjing First Hospital , Nanjing, China
| | - Yi Peng
- 2 Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University , Wuhan, China
| | - Li-ming Wang
- 1 Department of Cardiothoracic Surgery, Nanjing First Hospital , Nanjing, China
| | - Jian-kai Wang
- 1 Department of Cardiothoracic Surgery, Nanjing First Hospital , Nanjing, China
| | - Xin Chen
- 1 Department of Cardiothoracic Surgery, Nanjing First Hospital , Nanjing, China
| |
Collapse
|
22
|
Sphingosine kinase 1 activation enhances epidermal innate immunity through sphingosine-1-phosphate stimulation of cathelicidin production. J Dermatol Sci 2015; 79:229-34. [PMID: 26113114 DOI: 10.1016/j.jdermsci.2015.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/06/2015] [Accepted: 06/16/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND The ceramide metabolite, sphingosine-1-phosphate (S1P), regulates multiple cellular functions in keratinocytes (KC). We recently discovered that production of a key innate immune element, cathelicidin antimicrobial peptide (CAMP), is stimulated via a NF-κB-dependent mechanism that is activated by S1P when S1P is generated by sphingosine kinase (SPHK) 1. OBJECTIVE We investigated whether pharmacological modulation of SPHK1 activity, using a novel synthetic SPHK1 activator, (S)-methyl 2-(hexanamide)-3-(4-hydroxyphenyl) propanoate (MHP), stimulates CAMP expression. METHODS MHP-mediated changes in both S1P and CAMP downstream mediators were analyzed in normal cultured human KC by qRT-PCR, Western immunoblot, ELISA, confocal microscopy for immunohistochemistry, HPLC and ESI-LC/MS/MS, and microbial pathogen invasion/colonization in a human epidermal organotypic model. RESULTS Treatment with MHP directly activated SPHK1 and increased cellular S1P content in normal cultured human KC. Because MHP did not inhibit S1P lyase activity, which hydrolyses S1P, augumented S1P levels could be attributed to increased synthesis rather than blockade of S1P degradation. Next, we found that exogenous MHP significantly stimulated CAMP mRNA and protein production in KC, increases that were significantly suppressed by siRNA directed against SPHK1, but not by a scrambled control siRNA. NF-κB activation, assessed by nuclear translocation of NF-κB, occurred in cells following incubation with MHP. Conversely, pretreatment with a specific inhibitor of SPHK1 decreased MHP-induced nuclear translocation of NF-κB, and significantly attenuated the MHP-mediated increase in CAMP production. Finally, topical MHP significantly suppressed invasion of the virulent Staphylococcus aureus into murine skin explants. CONCLUSION MHP activation of SPHK1, a target enzyme of CAMP production, can stimulate innate immunity.
Collapse
|
23
|
Ji F, Mao L, Liu Y, Cao X, Xie Y, Wang S, Fei H. K6PC-5, a novel sphingosine kinase 1 (SphK1) activator, alleviates dexamethasone-induced damages to osteoblasts through activating SphK1-Akt signaling. Biochem Biophys Res Commun 2015; 458:568-575. [PMID: 25680461 DOI: 10.1016/j.bbrc.2015.02.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/02/2015] [Indexed: 02/01/2023]
Abstract
Long-term glucocorticoid usage is a common cause of non-traumatic femoral head osteonecrosis. Glucocorticoids (i.e. dexamethasone (Dex)) could directly induce damages to osteoblasts. In the current study, we investigated the potential activity of K6PC-5 [N-(1,3-dihydroxyisopropyl)-2-hexyl-3-oxo-decanamide], a novel sphingosine kinase 1 (SphK1) activator, against this process. Our data revealed that both osteoblastic-like MC3T3-E1 cells and primary murine osteoblasts were responsible to K6PC-5. K6PC-5 activated SphK1, increased sphingosine-1-phosphate (S1P) production and induced Akt phosphorylation in cultured osteoblasts. Functionally, K6PC-5 protected osteoblasts from Dex-induced apoptosis and necrosis. Such signaling and functional effects by K6PC-5 were prevented by the SphK1 inhibitor N,N-dimethylsphingosine (DMS), and by SphK1-siRNAs. On the other hand, exogenously-added S1P activated Akt and reduced Dex-induced osteoblast damages. LY294002 and MK-2206, two established Akt inhibitors, alleviated K6PC-5- or S1P-mediated osteoblast protection against Dex. Together, our results suggest that K6PC-5 alleviates Dex-induced osteoblast injuries through activating SphK1-Akt signaling. K6PC-5 might be further investigated in animal or clinical studies for its anti-glucocorticoids-associated osteonecrosis potential.
Collapse
Affiliation(s)
- Feng Ji
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Orthopedics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Li Mao
- Department of Endocrinology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Yuanyuan Liu
- Department of Endocrinology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Xiaojian Cao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yue Xie
- Department of Orthopedics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Shouguo Wang
- Department of Orthopedics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Haodong Fei
- Department of Orthopedics, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| |
Collapse
|
24
|
Schümann J, Grevot A, Ledieu D, Wolf A, Schubart A, Piaia A, Sutter E, Côté S, Beerli C, Pognan F, Billich A, Moulin P, Walker UJ. Reduced Activity of Sphingosine-1-Phosphate Lyase Induces Podocyte-related Glomerular Proteinuria, Skin Irritation, and Platelet Activation. Toxicol Pathol 2015; 43:694-703. [PMID: 25630683 DOI: 10.1177/0192623314565650] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Sphingosine-1-phosphate (S1P) lyase is considered as a drug target in autoimmune diseases based on the protective effect of reducing activity of the enzyme in animal models of inflammation. Since S1P lyase deficiency in mice causes a severe, lethal phenotype, it was of interest to investigate any pathological alterations associated with only partially reduced activity of S1P lyase as may be encountered upon pharmacological inhibition. Both genetic reduction of S1P lyase activity in mice and inhibition of S1P lyase with a low-molecular-weight compound in rats consistently resulted in podocyte-based kidney toxicity, which is the most severe finding. In addition, skin irritation and platelet activation were observed in both instances. The similarity of the findings in both the genetic model and the pharmacological study supports the value of analyzing inducible partially target-deficient mice for safety assessment. If the findings described in rodents translate to humans, target-related toxicity, particularly podocyte dysfunction, may limit chronic systemic treatment of autoimmune diseases with S1P lyase inhibitors. Furthermore, partial deficiency or inhibition of S1P lyase appears to provide an in vivo rodent model to enable studies on the mechanism of podocyte dysfunction.
Collapse
Affiliation(s)
- Jens Schümann
- Preclinical Safety, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Armelle Grevot
- Preclinical Safety, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - David Ledieu
- Preclinical Safety, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Armin Wolf
- Preclinical Safety, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Anna Schubart
- Autoimmunity, Transplantation, and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Alessandro Piaia
- Preclinical Safety, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Esther Sutter
- Preclinical Safety, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Serge Côté
- Preclinical Safety, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Christian Beerli
- Autoimmunity, Transplantation, and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - François Pognan
- Preclinical Safety, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Andreas Billich
- Autoimmunity, Transplantation, and Inflammation, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Pierre Moulin
- Preclinical Safety, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Ursula Junker Walker
- Preclinical Safety, Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
25
|
Bacakova M, Lopot F, Hadraba D, Varga M, Zaloudkova M, Stranska D, Suchy T, Bacakova L. Effects of fiber density and plasma modification of nanofibrous membranes on the adhesion and growth of HaCaT keratinocytes. J Biomater Appl 2014; 29:837-53. [DOI: 10.1177/0885328214546647] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
It may be possible to regulate the cell colonization of biodegradable polymer nanofibrous membranes by plasma treatment and by the density of the fibers. To test this hypothesis, nanofibrous membranes of different fiber densities were treated by oxygen plasma with a range of plasma power and exposure times. Scanning electron microscopy and mechanical tests showed significant modification of nanofibers after plasma treatment. The intensity of the fiber modification increased with plasma power and exposure time. The exposure time seemed to have a stronger effect on modifying the fiber. The mechanical behavior of the membranes was influenced by the plasma treatment, the fiber density, and their dry or wet state. Plasma treatment increased the membrane stiffness; however, the membranes became more brittle. Wet membranes displayed significantly lower stiffness than dry membranes. X-ray photoelectron spectroscopy (XPS) analysis showed a slight increase in oxygen-containing groups on the membrane surface after plasma treatment. Plasma treatment enhanced the adhesion and growth of HaCaT keratinocytes on nanofibrous membranes. The cells adhered and grew preferentially on membranes of lower fiber densities, probably due to the larger area of void spaces between the fibers.
Collapse
Affiliation(s)
- Marketa Bacakova
- Institute of Physiology, Academy of Sciences of the Czech Republic, Czech Republic
| | - Frantisek Lopot
- Dept. of Anatomy and Biomechanics, Faculty of Physical Education and Sport, Charles University, Czech Republic
| | - Daniel Hadraba
- Institute of Physiology, Academy of Sciences of the Czech Republic, Czech Republic
- Dept. of Anatomy and Biomechanics, Faculty of Physical Education and Sport, Charles University, Czech Republic
| | - Marian Varga
- Institute of Physics, Academy of Sciences of the Czech Republic, Czech Republic
| | - Margit Zaloudkova
- Institute of Rock Structure and Mechanics, Academy of Sciences of the Czech Republic, Czech Republic
| | | | - Tomas Suchy
- Institute of Rock Structure and Mechanics, Academy of Sciences of the Czech Republic, Czech Republic
| | - Lucie Bacakova
- Institute of Physiology, Academy of Sciences of the Czech Republic, Czech Republic
| |
Collapse
|
26
|
Kim B, Kim JE, Kim HS. Caffeic acid induces keratinocyte differentiation by activation of PPAR-α. J Pharm Pharmacol 2013; 66:84-92. [DOI: 10.1111/jphp.12159] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/16/2013] [Indexed: 01/27/2023]
Abstract
Abstract
Objectives
Peroxisome proliferator-activated receptors (PPAR)-α plays an important role in epidermal differentiation and barrier recovery, and topical treatment with PPAR-α agonists restores epidermal homeostasis in essential fatty acid deficiency and permeability barrier in skin disruptions. Therefore, we performed structure-based pharmacophore screening to search for a novel PPAR-α agonist. Caffeic acid was ultimately selected and evaluated for its effects on keratinocyte differentiation and epidermal permeability barrier.
Methods
The transactivation activity of PPAR-responsive element (PPRE) and cornified envelope (CE) formation were assayed. Also, immunoblot analysis and anti-oxidant activity were investigated on caffeic acid.
Key findings
Caffeic acid increases the transactivation activity of PPRE and CE formation in keratinocytes. In addition, caffeic acid promotes the expression of genes and proteins related to CE formation such as involucrin and transglutaminase-1. Additionally, anti-oxidant activity were improved by caffeic acid.
Conclusions
Caffeic acid can promote keratinocyte differentiation and restore skin barrier homeostasis and is suggested to be an appropriate skin therapeutic agent for improving epidermal permeability barrier function.
Collapse
Affiliation(s)
- Bora Kim
- R&D Center of Skin Science and Cosmetics, Enprani Co., Ltd, Incheon, Korea
| | - Jin Eun Kim
- R&D Center of Skin Science and Cosmetics, Enprani Co., Ltd, Incheon, Korea
| | - Hyun-Soo Kim
- Department of Food Science and Industry, Jungwon University, Chungbuk, Korea
| |
Collapse
|
27
|
Allende ML, Sipe LM, Tuymetova G, Wilson-Henjum KL, Chen W, Proia RL. Sphingosine-1-phosphate phosphatase 1 regulates keratinocyte differentiation and epidermal homeostasis. J Biol Chem 2013; 288:18381-91. [PMID: 23637227 DOI: 10.1074/jbc.m113.478420] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid whose levels are tightly regulated by its synthesis and degradation. Intracellularly, S1P is dephosphorylated by the actions of two S1P-specific phosphatases, sphingosine-1-phosphate phosphatases 1 and 2. To identify the physiological functions of S1P phosphatase 1, we have studied mice with its gene, Sgpp1, deleted. Sgpp1(-/-) mice appeared normal at birth, but during the 1st week of life they exhibited stunted growth and suffered desquamation, with most dying before weaning. Both Sgpp1(-/-) pups and surviving adults exhibited multiple epidermal abnormalities. Interestingly, the epidermal permeability barrier developed normally during embryogenesis in Sgpp1(-/-) mice. Keratinocytes isolated from the skin of Sgpp1(-/-) pups had increased intracellular S1P levels and displayed a gene expression profile that indicated overexpression of genes associated with keratinocyte differentiation. The results reveal S1P metabolism as a regulator of keratinocyte differentiation and epidermal homeostasis.
Collapse
Affiliation(s)
- Maria L Allende
- Genetics of Development and Disease Branch, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
28
|
Lopez-Pajares V, Yan K, Zarnegar BJ, Jameson KL, Khavari PA. Genetic pathways in disorders of epidermal differentiation. Trends Genet 2013; 29:31-40. [PMID: 23141808 PMCID: PMC5477429 DOI: 10.1016/j.tig.2012.10.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/02/2012] [Accepted: 10/04/2012] [Indexed: 10/27/2022]
Abstract
More than 100 human genetic skin diseases, impacting over 20% of the population, are characterized by disrupted epidermal differentiation. A significant proportion of the 90 genes identified in these disorders to date are concentrated within several functional pathways, suggesting the emergence of organizing themes in epidermal differentiation. Among these are the Notch, transforming growth factor β (TGFβ), IκB kinase (IKK), Ras/mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K), p63, and Wnt signaling pathways, as well as core biological processes mediating calcium homeostasis, tissue integrity, cornification, and lipid biogenesis. Here, we review recent results supporting the central role of these pathways in epidermal differentiation, highlighting the integration of genetic information with functional studies to illuminate the biological actions of these pathways in humans as well as to guide development of future therapeutics to correct their dysfunction.
Collapse
Affiliation(s)
| | - Karen Yan
- Program in Epithelial Biology, Stanford University, Stanford, CA 94305
| | - Brian J. Zarnegar
- Program in Epithelial Biology, Stanford University, Stanford, CA 94305
| | | | - Paul A. Khavari
- Program in Epithelial Biology, Stanford University, Stanford, CA 94305
| |
Collapse
|
29
|
Japtok L, Schaper K, Bäumer W, Radeke HH, Jeong SK, Kleuser B. Sphingosine 1-phosphate modulates antigen capture by murine Langerhans cells via the S1P2 receptor subtype. PLoS One 2012; 7:e49427. [PMID: 23145172 PMCID: PMC3493526 DOI: 10.1371/journal.pone.0049427] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 10/08/2012] [Indexed: 01/09/2023] Open
Abstract
Dendritic cells (DCs) play a pivotal role in the development of cutaneous contact hypersensitivity (CHS) and atopic dermatitis as they capture and process antigen and present it to T lymphocytes in the lymphoid organs. Recently, it has been indicated that a topical application of the sphingolipid sphingosine 1-phosphate (S1P) prevents the inflammatory response in CHS, but the molecular mechanism is not fully elucidated. Here we indicate that treatment of mice with S1P is connected with an impaired antigen uptake by Langerhans cells (LCs), the initial step of CHS. Most of the known actions of S1P are mediated by a family of five specific G protein-coupled receptors. Our results indicate that S1P inhibits macropinocytosis of the murine LC line XS52 via S1P2 receptor stimulation followed by a reduced phosphatidylinositol 3-kinase (PI3K) activity. As down-regulation of S1P2 not only diminished S1P-mediated action but also enhanced the basal activity of LCs on antigen capture, an autocrine action of S1P has been assumed. Actually, S1P is continuously produced by LCs and secreted via the ATP binding cassette transporter ABCC1 to the extracellular environment. Consequently, inhibition of ABCC1, which decreased extracellular S1P levels, markedly increased the antigen uptake by LCs. Moreover, stimulation of sphingosine kinase activity, the crucial enzyme for S1P formation, is connected not only with enhanced S1P levels but also with diminished antigen capture. These results indicate that S1P is essential in LC homeostasis and influences skin immunity. This is of importance as previous reports suggested an alteration of S1P levels in atopic skin lesions.
Collapse
Affiliation(s)
- Lukasz Japtok
- Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany
| | - Katrin Schaper
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Wolfgang Bäumer
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Heinfried H. Radeke
- Pharmazentrum Frankfurt, Clinic of the Johann Wolfgang Goethe-University, Frankfurt/Main, Germany
| | | | - Burkhard Kleuser
- Faculty of Mathematics and Natural Science, Institute of Nutritional Science, Department of Toxicology, University of Potsdam, Potsdam, Germany
- * E-mail:
| |
Collapse
|
30
|
Kendall AC, Nicolaou A. Bioactive lipid mediators in skin inflammation and immunity. Prog Lipid Res 2012; 52:141-64. [PMID: 23124022 DOI: 10.1016/j.plipres.2012.10.003] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 10/15/2012] [Accepted: 10/17/2012] [Indexed: 12/20/2022]
Abstract
The skin is the primary barrier from the outside environment, protecting the host from injury, infectious pathogens, water loss and solar ultraviolet radiation. In this role, it is supported by a highly organized system comprising elements of innate and adaptive immunity, responsive to inflammatory stimuli. The cutaneous immune system is regulated by mediators such as cytokines and bioactive lipids that can initiate rapid immune responses with controlled inflammation, followed by efficient resolution. However, when immune responses are inadequate or mounted against non-infectious agents, these mediators contribute to skin pathologies involving unresolved or chronic inflammation. Skin is characterized by active lipid metabolism and fatty acids play crucial roles both in terms of structural integrity and functionality, in particular when transformed to bioactive mediators. Eicosanoids, endocannabinoids and sphingolipids are such key bioactive lipids, intimately involved in skin biology, inflammation and immunity. We discuss their origins, role and influence over various cells of the epidermis, dermis and cutaneous immune system and examine their function in examples of inflammatory skin conditions. We focus on psoriasis, atopic and contact dermatitis, acne vulgaris, wound healing and photodermatology that demonstrate dysregulation of bioactive lipid metabolism and examine ways of using this insight to inform novel therapeutics.
Collapse
Affiliation(s)
- Alexandra C Kendall
- School of Pharmacy and Centre for Skin Sciences, School of Life Sciences, University of Bradford, Richmond Road, Bradford BD7 1DP, UK
| | | |
Collapse
|
31
|
SERCA2-controlled Ca²+-dependent keratinocyte adhesion and differentiation is mediated via the sphingolipid pathway: a therapeutic target for Darier's disease. J Invest Dermatol 2012; 132:1188-95. [PMID: 22277942 PMCID: PMC3305850 DOI: 10.1038/jid.2011.447] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Darier’s Disease (DD), caused by mutations in the endoplasmic reticulum (ER) Ca2+ ATPase ATP2A2 (SERCA2b), is a skin disease that exhibits impaired epidermal cell-to-cell adhesion and altered differentiation. Although previous studies have shown that keratinocyte Ca2+ sequestration and fluxes are controlled by sphingolipid signaling, the role of this signaling pathway in DD previously has not been investigated. We show here that sphingosine levels increase and sphingosine kinase (SPHK1) expression decreases after inactivating SERCA2b with the specific SERCA2 inhibitors thapsigargin (TG) or siRNA to SERCA2b. Conversely, inhibiting sphingosine lyase rescues the defects in keratinocyte differentiation, E-cadherin localization, Desmoplakin (DP) translocation, and ER Ca2+ sequestration seen in TG-treated keratinocytes. To our knowledge, it was previously unreported that the keratinocyte sphingolipid and Ca2+ signaling pathways intersect in ATP2A2- controlled ER Ca2+ sequestration, E-cadherin and desmoplakin localization and Ca2+ - controlled differentiation, and thus may be important mediators in DD.
Collapse
|
32
|
Singh AT, Dharmarajan A, Aye ILMH, Keelan JA. Sphingosine-sphingosine-1-phosphate pathway regulates trophoblast differentiation and syncytialization. Reprod Biomed Online 2011; 24:224-34. [PMID: 22197131 DOI: 10.1016/j.rbmo.2011.10.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 10/25/2011] [Accepted: 10/26/2011] [Indexed: 12/30/2022]
Abstract
Sphingosine and sphingosine-1-phosphate (S1P) are involved in regulating cell differentiation. This study postulated that changes in sphingolipid biosynthesis and metabolism are important in trophoblast syncytialization and therefore examined the production, metabolism and actions of sphingosine and S1P during spontaneous trophoblast differentiation and fusion in vitro. Significant declines in intracellular sphingosine concentration (P≤0.05) and sphingosine kinase 1 (SPHK1) expression (P≤0.01) were observed during trophoblast syncytialization. Secreted S1P concentrations dropped steeply after 72h, before rising to basal concentrations with syncytialization. Intracellular S1P concentrations were undetectable throughout. Treating cells with exogenous sphingosine (P≤0.01), S1P (P≤0.001) or a specific SPHK1 inhibitor (P≤0.05) for up to 72h in culture significantly inhibited trophoblast differentiation (measured as reduced human chorionic gonadotrophin production); effects on other biochemical and morphological markers of differentiation were absent or inconsistent. Phosphorylation of Akt, an established down-stream target of S1P that spontaneously declines with trophoblast differentiation, was markedly reduced by S1P (P≤0.05). In conclusion, changes in the sphingosine-S1P pathway are involved in the regulation of trophoblast differentiation in term human placenta. Dysregulation of sphingolipid homeostasis could, therefore, disrupt placental formation and function with deleterious consequences for pregnancy outcome.
Collapse
Affiliation(s)
- Ambika T Singh
- School of Women's and Infants' Health, The University of Western Australia, Subiaco, Perth, Western Australia, Australia
| | | | | | | |
Collapse
|
33
|
Kim J, Yun H, Cho Y. Analysis of ceramide metabolites in differentiating epidermal keratinocytes treated with calcium or vitamin C. Nutr Res Pract 2011; 5:396-403. [PMID: 22125676 PMCID: PMC3221824 DOI: 10.4162/nrp.2011.5.5.396] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/17/2011] [Accepted: 09/22/2011] [Indexed: 01/07/2023] Open
Abstract
Ceramides (Cer) comprise the major constituent of sphingolipids in the epidermis and are known to play diverse roles in the outermost layers of the skin including water retention and provision of a physical barrier. In addition, they can be hydrolyzed into free sphingoid bases such as C18 sphingosine (SO) and C18 sphinganine (SA) or can be further metabolized to C18 So-1-phosphate (S1P) and C18 Sa-1-phosphate (Sa1P) in keratinocytes. The significance of ceramide metabolites emerged from studies reporting altered levels of SO and SA in skin disorders and the role of S1P and Sa1P as signaling lipids. However, the overall metabolism of sphingoid bases and their phosphates during keratinocyte differentiation remains not fully understood. Therefore, in this study, we analyzed these Cer metabolites in the process of keratinocyte differentiation. Three distinct keratinocyte differentiation stages were prepared using 0.07 mM calcium (Ca2+) (proliferation stage), 1.2 mM Ca2+ (early differentiation stage) in serum-free medium, or serum-containing medium with vitamin C (50 µL/mL) (late differentiation stage). Serum-containing medium was also used to determine whether vitamin C increases the concentrations of sphingoid bases and their phosphates. The production of sphingoid bases and their phosphates after hydrolysis by alkaline phosphatase was determined using high-performance liquid chromatography. Compared to cells treated with 0.07 mM Ca2+, levels of SO, SA, S1P, and SA1P were not altered after treatment with 1.2 mM Ca2+. However, in keratinocytes cultured in serum-containing medium with vitamin C, levels of SO, SA, S1P, and SA1P were dramatically higher than those in 0.07- and 1.2-mM Ca2+-treated cells; however, compared to serum-containing medium alone, vitamin C did not significantly enhance their production. Taken together, we demonstrate that late differentiation induced by vitamin C and serum was accompanied by dramatic increases in the concentration of sphingoid bases and their phosphates, although vitamin C alone had no effect on their production.
Collapse
Affiliation(s)
- Juyoung Kim
- Department of Medical Nutrition, Graduate School of East-West Medical Science, Kyung Hee University, Seocheon-dong, Giheung-gu, Yongin-si, Gyeonggi 446-701, Korea
| | | | | |
Collapse
|
34
|
The receptor tyrosine kinase FGFR2b/KGFR controls early differentiation of human keratinocytes. PLoS One 2011; 6:e24194. [PMID: 21957444 PMCID: PMC3177842 DOI: 10.1371/journal.pone.0024194] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 08/04/2011] [Indexed: 12/23/2022] Open
Abstract
The FGFRs trigger divergent responses, such as proliferation and differentiation, and the cell type as well as the context-dependent signaling are crucial for the functional outcome. The FGFR2b/KGFR is expressed exclusively on epithelial cells and plays a key role in skin homeostasis. Here we analyzed in vitro the role of KGFR in the early differentiation of keratinocytes modulating its expression by KGFR cDNA transient transfection or KGFR siRNA microinjection and inducing a synchronous wave of differentiation in pre-confluent cells. Immunofluorescence, biochemical and molecular approaches demonstrated that KGFR overexpression increased the early differentiation marker keratin 1 at both transcriptional and translational levels, while receptor depletion reduced it. Ligand-dependent receptor activation and signaling were required for this differentiative effect. Overexpression of kinase negative KGFR mutant or Tyr769 KGFR signaling mutant, which is not able to recruit and activate PLC-γ, showed that the receptor kinase activity, but not its PLCγ-mediated signaling, is required for differentiation. Reduction of K1 expression, obtained by AKT inhibition, demonstrated that the PI3K/Akt signaling pathway is involved in the control of KGFR-mediated keratinocyte differentiation. This in vitro experimental model indicates that FGFR2b/KGFR expression represents a key event regulating keratinocyte early differentiation during the switch from undifferentiated to differentiating cells.
Collapse
|
35
|
Bernacchioni C, Cencetti F, Kwon MJ, Gwak HS, Jeong SK, Bruni P, Donati C. The sphingosine kinase activator K6PC-5 stimulates C2C12 myoblast differentiation. Int J Immunopathol Pharmacol 2011; 24:55-62. [PMID: 21496387 DOI: 10.1177/039463201102400107] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Previously, K6PC-5, a synthetic derivative of ceramide, was demonstrated to activate sphingosine kinase (SK)-1 in keratinocytes. In this study its potential biological effect in mouse myoblasts was examined. The obtained results show that K6PC-5 promotes myogenic differentiation by enhancing myogenic marker expression, differentiation index and fusion index. Interestingly, its biological action was prevented by pharmacological inhibition of SK or S1P2 receptor, in full agreement with their recognized role in myoblast differentiation. This is the first evidence that pharmacological activation of SK accelerates myogenesis and suggests that this new therapeutic strategy could be possibly employed in skeletal muscle disorders where muscle regeneration is deficient.
Collapse
Affiliation(s)
- C Bernacchioni
- Dipartimento di Scienze Biochimiche, Università di Firenze; Istituto Interuniversitario di Miologia, Firenze, Italy
| | | | | | | | | | | | | |
Collapse
|
36
|
Sugita K, Kabashima K, Sakabe JI, Yoshiki R, Tanizaki H, Tokura Y. FTY720 regulates bone marrow egress of eosinophils and modulates late-phase skin reaction in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1881-7. [PMID: 20802177 DOI: 10.2353/ajpath.2010.100119] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Eosinophilia in the blood and skin is frequently observed in patients with certain inflammatory skin diseases, such as atopic dermatitis. However, the mechanism underlying eosinophil circulation and the role of eosinophils in cutaneous immune responses remain unclear. In repeated hapten application-induced cutaneous responses in BALB/c mice, the administration of FTY720 before the last challenge decreased the number of skin-infiltrating eosinophils and reduced the late-phase reaction. A similar reduction of the late-phase reaction was observed by a sphingosine-1-phosphate G protein-coupled receptor (S1P1)-selective agonist, SEW2871. We monitored numerous alterations of eosinophils in the blood, spleen, bone marrow, and lymph nodes of interleukin-5 transgenic mice, used as an eosinophilia model, following FTY720 administration. The number of circulating eosinophils was significantly decreased after treatment with FTY720, and eosinophils accumulated in the bone marrow. In addition, eosinophils expressed S1P1, S1P3, and S1P4 mRNAs, and their chemotactic response to S1P was abolished by FTY720 as well as by SEW2871. These findings suggest that FTY720 affects the number of eosinophils in both the blood and skin by inhibiting the egress of eosinophils from the bone marrow and thus downmodulating the late-phase reaction.
Collapse
Affiliation(s)
- Kazunari Sugita
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan.
| | | | | | | | | | | |
Collapse
|
37
|
Facchinetti MM, Gandini NA, Fermento ME, Sterin-Speziale NB, Ji Y, Patel V, Gutkind JS, Rivadulla MG, Curino AC. The expression of sphingosine kinase-1 in head and neck carcinoma. Cells Tissues Organs 2010; 192:314-24. [PMID: 20606403 DOI: 10.1159/000318173] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2010] [Indexed: 12/22/2022] Open
Abstract
Sphingosine kinase-1 (SPHK1) modulates the proliferation, apoptosis and differentiation of keratinocytes through the regulation of ceramide and sphingosine-1-phosphate levels. However, studies on the expression of SPHK1 in human head and neck squamous cell carcinoma (HNSCC) specimens are lacking. Therefore, the aim of the present work was to evaluate SPHK1 expression in human primary HNSCCs and to correlate the results with clinical and anatomopathological parameters. We investigated the expression of this protein by immunohistochemistry performed in tissue microarrays of HNSCC and in an independent cohort of 37 paraffin-embedded specimens. SPHK1 expression was further validated by real-time PCR performed on laser capture-microdissected tissue samples. The positive rate of SPHK1 protein in the cancerous tissues was significantly higher (74%) than that in the nontumor oral tissues (23%), and malignant tissues showed stronger immunoreactivity for SPHK1 than normal matching samples. These results were confirmed by real-time PCR quantification of SPHK1 mRNA. Interestingly, the positive expression of SPHK1 was associated with shorter patient survival time (Kaplan-Meier survival curves) and with the loss of p21 expression. Taken together, these results demonstrate that SPHK1 is upregulated in HNSCC and provide clues of the role SPHK1 might play in tumor progression.
Collapse
Affiliation(s)
- María M Facchinetti
- Instituto de Investigaciones Bioquímicas Bahía Blanca, INIBIBB-CONICET, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Topical application of sphingosine-1-phosphate and FTY720 attenuate allergic contact dermatitis reaction through inhibition of dendritic cell migration. J Invest Dermatol 2009; 129:1954-62. [PMID: 19194476 DOI: 10.1038/jid.2008.454] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Migration of Langerhans cells (LCs) from the skin to the lymph node is an essential step in the pathogenesis of allergic contact dermatitis (ACD). Therefore, inhibition of LC-migration could be a promising strategy to improve this skin disease. Effects of sphingosine-1-phosphate (S1P) and the immunomodulator FTY720 on LC trafficking is not well defined, yet. Thus, we investigated the action of topically administered S1P and FTY720 in a murine model of ACD. Most interestingly, FTY720 as well as S1P inhibited the inflammatory reaction in the elicitation phase of ACD. In the sensitization phase, FTY720, and S1P reduced the weight and cell count of the draining auricular lymph node, as well as immigrated dendritic cells provoked by repetitive topical administration of the hapten. Correspondingly, the density of LCs in the epidermis was higher in FTY720- and S1P-treated mice compared to vehicle treatment. A skin dendritic cell migration assay confirmed the significant inhibition of dendritic cell migration by FTY720 and S1P. These data supply conclusive evidence that the strategy of targeting the migratory response of LCs with locally acting S1P or FTY720 represents an emerging option in the treatment of allergic skin diseases like contact hypersensitivity and atopic dermatitis.
Collapse
|
39
|
K6PC-5, a sphingosine kinase activator, induces anti-aging effects in intrinsically aged skin through intracellular Ca2+ signaling. J Dermatol Sci 2008; 51:89-102. [PMID: 18420384 DOI: 10.1016/j.jdermsci.2008.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Revised: 02/20/2008] [Accepted: 03/03/2008] [Indexed: 01/01/2023]
Abstract
BACKGROUND Sphingosine-1-phosphate (S1P), a bioactive sphingolipid metabolite, regulates multiple cellular responses such as Ca(2+) signaling, growth, survival, and differentiation. Because sphingosine kinase (SK) is the enzyme directly responsible for the production of S1P, many factors have been identified that regulate its activity and subsequent S1P levels. To date, there are no reports to demonstrate a chemically induced, direct activation of SK. OBJECTIVE Here we have studied the effects of K6PC-5 as a newly synthesized SK activator on fibroblast proliferation in both human fibroblasts and aged mouse skin. To demonstrate that K6PC-5 has S1P-mediated action mechanism in fibroblasts, we have measured SK-dependent intracellular Ca(2+) signaling. METHODS Fibroblasts were cultured primarily from human foreskin and were used to study the effect of K6PC-5 and S1P on intracellular Ca(2+) signaling and fibroblast proliferation. Changes in intracellular Ca(2+) were detected by fluorescence with fura-2/AM. To study skin anti-aging effects of K6PC-5, we used intrinsically aged hairless mice (56 weeks old). RESULTS K6PC-5 promoted fibroblast proliferation and procollagen production in human fibroblasts significantly. K6PC-5 induced intracellular Ca(2+) concentration ([Ca(2+)](i)) oscillations in human fibroblasts. Both dimethylsphingosine and dihydroxysphingosine, SK inhibitors, and the transfection of SK1-siRNA blocked the K6PC-5-induced increases in [Ca(2+)](i), an effect independent of the classical PLC/IP(3)-mediated pathway. The K6PC-5-induced [Ca(2+)](i) oscillations were dependent on thapsigargin-sensitive Ca(2+) stores and Ca(2+) entry. Topical application of K6PC-5 for 2 weeks to intrinsically aged hairless mice enhanced fibroblast proliferation, collagen production, and eventually increased dermal thickness (10%). K6PC-5 also promoted specific epidermal differentiation marker proteins, including involucrin, loricrin, filaggrin, and keratin 5, without any alterations on epidermal barrier function. CONCLUSION These results suggest that K6PC-5 acts to regulate fibroblast proliferation through intracellular S1P production, and can further promote keratinocyte differentiation. We anticipate that the regulation of S1P levels may represent a novel approach for the treatment of skin disorders, including skin aging.
Collapse
|
40
|
Park HY, Youm JK, Kwon MJ, Park BD, Lee SH, Choi EH. K6PC-5, a novel sphingosine kinase activator, improves long-term ultraviolet light-exposed aged murine skin. Exp Dermatol 2008; 17:829-36. [PMID: 18341573 DOI: 10.1111/j.1600-0625.2008.00708.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Sphingosine-1-phosphate (S1P), which is formed by phosphorylation of sphingosine through a process catalysed by sphingosine kinase (SK), is a multifunctional mediator of a variety of cellular responses including proliferation, differentiation, motility, and survival. K6PC-5, which was recently synthesized as a novel SK activator, is expected to increase S1P levels. Indeed studies have already demonstrated that K6PC-5 exhibits anti-aging effects on intrinsic aged murine skin by increasing fibroblasts, collagen synthesis, dermal thickness, and epidermal differentiation. However, photoaging and intrinsic aging have highly different clinical and histopathological properties. In this study, we developed a photoaged murine model by exposing mice that were 56 weeks old to ultraviolet (UV)B and UVA radiation for 8 weeks. We then investigated whether K6PC-5, as an SK activator, had anti-aging effects on photoaged murine skin in addition to its effects on intrinsic aged murine skin and determined the mechanism. K6PC-5 increased dermal collagen density in photoaged skin through increases in fibroblasts and collagen production. Photoaged murine skin treated with K6PC-5 showed an increase in stratum corneum (SC) integrity with increased corneodesmosome density and an improvement in barrier recovery rate. Matrix metalloproteinase 13 remained unchanged. These results indicate that topical application of K6PC-5 improves photoaged skin by improving skin barrier and increasing fibroblast count and function. In conclusion, K6PC-5, as an S1P activator, improves long-term UV-exposed aged skin as well as intrinsic aged skin.
Collapse
Affiliation(s)
- Hwa-young Park
- Department of Dermatology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | | | | | | | | | | |
Collapse
|