1
|
Banerjee R, Knauer LA, Iyer D, Barlow SE, Shalaby H, Dehghan R, Scallan JP, Yang Y. Rictor, an mTORC2 Protein, Regulates Murine Lymphatic Valve Formation Through the AKT-FOXO1 Signaling. Arterioscler Thromb Vasc Biol 2024; 44:2004-2023. [PMID: 39087350 PMCID: PMC11335088 DOI: 10.1161/atvbaha.124.321164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Lymphatic valves are specialized structures in collecting lymphatic vessels and are crucial for preventing retrograde lymph flow. Mutations in valve-forming genes have been clinically implicated in the pathology of congenital lymphedema. Lymphatic valves form when oscillatory shear stress from lymph flow signals through the PI3K/AKT pathway to promote the transcription of valve-forming genes that trigger the growth and maintenance of lymphatic valves. Conventionally, in many cell types, AKT is phosphorylated at Ser473 by the mTORC2 (mammalian target of rapamycin complex 2). However, mTORC2 has not yet been implicated in lymphatic valve formation. METHODS In vivo and in vitro techniques were used to investigate the role of Rictor, a critical component of mTORC2, in lymphatic endothelium. RESULTS Here, we showed that embryonic and postnatal lymphatic deletion of Rictor, a critical component of mTORC2, led to a significant decrease in lymphatic valves and prevented the maturation of collecting lymphatic vessels. RICTOR knockdown in human dermal lymphatic endothelial cells not only reduced the level of activated AKT and the expression of valve-forming genes under no-flow conditions but also abolished the upregulation of AKT activity and valve-forming genes in response to oscillatory shear stress. We further showed that the AKT target, FOXO1 (forkhead box protein O1), a repressor of lymphatic valve formation, had increased nuclear activity in Rictor knockout mesenteric lymphatic endothelial cells in vivo. Deletion of Foxo1 in Rictor knockout mice restored the number of valves to control levels in lymphatic vessels of the ear and mesentery. CONCLUSIONS Our work identifies a novel role for RICTOR in the mechanotransduction signaling pathway, wherein it activates AKT and prevents the nuclear accumulation of the valve repressor, FOXO1, which ultimately enables the formation and maintenance of lymphatic valves.
Collapse
Affiliation(s)
- Richa Banerjee
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Luz A. Knauer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Drishya Iyer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Sara E. Barlow
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Hanan Shalaby
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Razieh Dehghan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Joshua P. Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| |
Collapse
|
2
|
Oliveira-Paula GH, Liu S, Maira A, Ressa G, Ferreira GC, Quintar A, Jayakumar S, Almonte V, Parikh D, Valenta T, Basler K, Hla T, Riascos-Bernal DF, Sibinga NES. The β-catenin C terminus links Wnt and sphingosine-1-phosphate signaling pathways to promote vascular remodeling and atherosclerosis. SCIENCE ADVANCES 2024; 10:eadg9278. [PMID: 38478616 PMCID: PMC10936954 DOI: 10.1126/sciadv.adg9278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 02/07/2024] [Indexed: 03/17/2024]
Abstract
Canonical Wnt and sphingosine-1-phosphate (S1P) signaling pathways are highly conserved systems that contribute to normal vertebrate development, with key consequences for immune, nervous, and cardiovascular system function; despite these functional overlaps, little is known about Wnt/β-catenin-S1P cross-talk. In the vascular system, both Wnt/β-catenin and S1P signals affect vessel maturation, stability, and barrier function, but information regarding their potential coordination is scant. We report an instance of functional interaction between the two pathways, including evidence that S1P receptor 1 (S1PR1) is a transcriptional target of β-catenin. By studying vascular smooth muscle cells and arterial injury response, we find a specific requirement for the β-catenin carboxyl terminus, which acts to induce S1PR1, and show that this interaction is essential for vascular remodeling. We also report that pharmacological inhibition of the β-catenin carboxyl terminus reduces S1PR1 expression, neointima formation, and atherosclerosis. These findings provide mechanistic understanding of how Wnt/β-catenin and S1P systems collaborate during vascular remodeling and inform strategies for therapeutic manipulation.
Collapse
Affiliation(s)
- Gustavo H. Oliveira-Paula
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sophia Liu
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alishba Maira
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gaia Ressa
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Graziele C. Ferreira
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Amado Quintar
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Smitha Jayakumar
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Vanessa Almonte
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dippal Parikh
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tomas Valenta
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Konrad Basler
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Dario F. Riascos-Bernal
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nicholas E. S. Sibinga
- Department of Medicine (Cardiology Division), Department of Developmental and Molecular Biology, and Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
3
|
Uranbileg B, Sakai E, Kubota M, Isago H, Sumitani M, Yatomi Y, Kurano M. Development of an advanced liquid chromatography-tandem mass spectrometry measurement system for simultaneous sphingolipid analysis. Sci Rep 2024; 14:5699. [PMID: 38459112 PMCID: PMC10923881 DOI: 10.1038/s41598-024-56321-w] [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: 11/21/2023] [Accepted: 03/05/2024] [Indexed: 03/10/2024] Open
Abstract
Mass spectrometry-based lipidomics approaches offer valuable tools for the detection and quantification of various lipid species, including sphingolipids. The present study aimed to develop a new method to simultaneously detect various sphingolipid species that applies to diverse biological samples. We developed and validated a measurement system by employing a single-column liquid chromatography-mass spectrometry system utilizing a normal-phase separation mode with positive ionization. The measurement system provided precision with a coefficient of variant below 20% for sphingolipids in all types of samples, and we observed good linearity in diluted serum samples. This system can measure the following sphingolipids: sphingosine 1-phosphate (S1P), sphingosine (Sph), dihydroS1P (dhS1P), dihydroSph (dhSph), ceramide 1-phosphate (Cer1P), hexosylceramide (HexCer), lactosylceramide (LacCer), dh-ceramide, deoxy-ceramide, deoxy-dh-ceramide, and sphingomyelin (SM). By measuring these sphingolipids in cell lysates where S1P lyase expression level was modulated, we could observe significant and dynamic modulations of sphingolipids in a comprehensive manner. Our newly established and validated measurement system can simultaneously measure many kinds of sphingolipids in biological samples. It holds great promise as a valuable tool for laboratory testing applications to detect overall modulations of sphingolipids, which have been proposed to be involved in pathogenesis processes in a series of elegant basic research studies.
Collapse
Affiliation(s)
- Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Eri Sakai
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Nihon Waters K.K., Tokyo, Japan
| | | | - Hideaki Isago
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masahiko Sumitani
- Department of Pain and Palliative Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
4
|
Yang S, Li HW, Tian JY, Wang ZK, Chen Y, Zhan TT, Ma CY, Feng M, Cao SF, Zhao Y, Li X, Ren J, Liu Q, Jin LY, Wang ZQ, Jiang WY, Zhao YX, Zhang Y, Liu X. Myeloid-derived growth factor suppresses VSMC dedifferentiation and attenuates postinjury neointimal formation in rats by activating S1PR2 and its downstream signaling. Acta Pharmacol Sin 2024; 45:98-111. [PMID: 37726422 PMCID: PMC10770085 DOI: 10.1038/s41401-023-01155-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/13/2023] [Indexed: 09/21/2023] Open
Abstract
Restenosis after angioplasty is caused usually by neointima formation characterized by aberrant vascular smooth muscle cell (VSMC) dedifferentiation. Myeloid-derived growth factor (MYDGF), secreted from bone marrow-derived monocytes and macrophages, has been found to have cardioprotective effects. In this study we investigated the effect of MYDGF to postinjury neointimal formation and the underlying mechanisms. Rat carotid arteries balloon-injured model was established. We found that plasma MYDGF content and the level of MYDGF in injured arteries were significantly decreased after balloon injury. Local application of exogenous MYDGF (50 μg/mL) around the injured vessel during balloon injury markedly ameliorated the development of neointimal formation evidenced by relieving the narrow endovascular diameter, improving hemodynamics, and reducing collagen deposition. In addition, local application of MYDGF inhibited VSMC dedifferentiation, which was proved by reversing the elevated levels of osteopontin (OPN) protein and decreased levels of α-smooth muscle actin (α-SMA) in the left carotid arteries. We showed that PDGF-BB (30 ng/mL) stimulated VSMC proliferation, migration and dedifferentiation in vitro; pretreatment with MYDGF (50-200 ng/mL) concentration-dependently eliminated PDGF-BB-induced cell proliferation, migration and dedifferentiation. Molecular docking revealed that MYDGF had the potential to bind with sphingosine-1-phosphate receptor 2 (S1PR2), which was confirmed by SPR assay and Co-IP analysis. Pretreatment with CCG-1423 (Rho signaling inhibitor), JTE-013 (S1PR2 antagonist) or Ripasudil (ROCK inhibitor) circumvented the inhibitory effects of MYDGF on VSMC phenotypic switching through inhibiting S1PR2 or its downstream RhoA-actin monomers (G-actin) /actin filaments (F-actin)-MRTF-A signaling. In summary, this study proves that MYDGF relieves neointimal formation of carotid arteries in response to balloon injury in rats, and suppresses VSMC dedifferentiation induced by PDGF-BB via S1PR2-RhoA-G/F-actin-MRTF-A signaling pathway. In addition, our results provide evidence for cross talk between bone marrow and vasculature.
Collapse
Affiliation(s)
- Shuang Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Hou-Wei Li
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jia-Ying Tian
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Zheng-Kai Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Yi Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Ting-Ting Zhan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Chun-Yue Ma
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Min Feng
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Shi-Feng Cao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Yu Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Xue Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Jing Ren
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Qian Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Lu-Ying Jin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Zhi-Qi Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Wen-Yu Jiang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Yi-Xiu Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Yan Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China.
| | - Xue Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China.
| |
Collapse
|
5
|
Wang W, Zhao Y, Zhu G. The role of sphingosine-1-phosphate in the development and progression of Parkinson's disease. Front Cell Neurosci 2023; 17:1288437. [PMID: 38179204 PMCID: PMC10764561 DOI: 10.3389/fncel.2023.1288437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Parkinson's disease (PD) could be viewed as a proteinopathy caused by changes in lipids, whereby modifications in lipid metabolism may lead to protein alterations, such as the accumulation of alpha-synuclein (α-syn), ultimately resulting in neurodegeneration. Although the loss of dopaminergic neurons in the substantia nigra is the major clinical manifestation of PD, the etiology of it is largely unknown. Increasing evidence has highlighted the important role of lipids in the pathophysiology of PD. Sphingosine-1-phosphate (S1P), a signaling lipid, has been suggested to have a potential association with the advancement and worsening of PD. Therefore, better understanding the mechanisms and regulatory proteins is of high interest. Most interestingly, S1P appears to be an important target to offers a new strategy for the diagnosis and treatment of PD. In this review, we first introduce the basic situation of S1P structure, function and regulation, with a special focus on the several pathways. We then briefly describe the regulation of S1P signaling pathway on cells and make a special focused on the cell growth, proliferation and apoptosis, etc. Finally, we discuss the function of S1P as potential therapeutic target to improve the clinical symptoms of PD, and even prevent the progression of the PD. In the context of PD, the functions of S1P modulators have been extensively elucidated. In conclusion, S1P modulators represent a novel and promising therapeutic principle and therapeutic method for PD. However, more research is required before these drugs can be considered as a standard treatment option for PD.
Collapse
Affiliation(s)
- Wang Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guoxue Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
6
|
Banerjee R, Knauer LA, Iyer D, Barlow SE, Scallan JP, Yang Y. Rictor induces AKT signaling to regulate lymphatic valve formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544698. [PMID: 37397997 PMCID: PMC10312634 DOI: 10.1101/2023.06.12.544698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Lymphatic valves are specialized structures of the collecting lymphatic vessels and are crucial for preventing retrograde lymph flow. Mutations in valve-forming genes have been clinically implicated in the pathology of congenital lymphedema. Lymphatic valves form when oscillatory shear stress (OSS) from lymph flow signals through the PI3K/AKT pathway to promote the transcription of valve-forming genes that trigger the growth and maintenance of lymphatic valves throughout life. Conventionally, in other tissue types, AKT activation requires dual kinase activity and the mammalian target of rapamycin complex 2 (mTORC2) commands this process by phosphorylating AKT at Ser473. Here we showed that embryonic and postnatal lymphatic deletion of Rictor , a critical component of mTORC2, led to a significant decrease in lymphatic valves and prevented the maturation of collecting lymphatic vessels. RICTOR knockdown in human lymphatic endothelial cells (hdLECs) not only significantly reduced the level of activated AKT and the expression of valve-forming genes under no-flow conditions, but also abolished the upregulation of AKT activity and valve-forming genes in response to flow. We further showed that the AKT target, FOXO1, a repressor of lymphatic valve formation, had increased nuclear activity in Rictor knockout mesenteric LECs, in vivo . Deletion of Foxo1 in Rictor knockout mice restored the number of valves to control levels in both mesenteric and ear lymphatics. Our work revealed a novel role of RICTOR signaling in the mechanotransduction signaling pathway, wherein it activates AKT and prevents the nuclear accumulation of the valve repressor, FOXO1, which ultimately allows the formation and maintenance of a normal lymphatic valve.
Collapse
|
7
|
Pan D, Wu W, Zuo G, Xie X, Li H, Ren X, Kong C, Zhou W, Zhang Z, Waterfall M, Chen S. Sphingosine 1-phosphate receptor 2 promotes erythrocyte clearance by vascular smooth muscle cells in intraplaque hemorrhage through MFG-E8 production. Cell Signal 2022; 98:110419. [PMID: 35905868 DOI: 10.1016/j.cellsig.2022.110419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/13/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
Intraplaque hemorrhage (IPH) accelerates atherosclerosis progression. To scavenge excessive red blood cells (RBCs), vascular smooth muscle cells (VSMCs) with great plasticity may function as phagocytes. Here, we investigated the erythrophagocytosis function of VSMCs and possible regulations involved. Based on transcriptional microarray analysis, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis showed that genes up-regulated in human carotid atheroma with IPH were enriched in functions of phagocytic activities, while those down-regulated were enriched in VSMCs contraction function. Transcriptional expression of Milk fat globule-epidermal growth factor 8 (MFG-E8) was also down-regulated in atheroma with IPH. In high-fat diet-fed apolipoprotein E-deficient mice, erythrocytes were present in cells expressing VSMC markers αSMA in the brachiocephalic artery, suggesting VSMCs play a role in erythrophagocytosis. Using immunofluorescence and flow cytometry, we also found that eryptotic RBCs were bound to and internalized by VSMCs in a phosphatidylserine/MFG-E8/integrin αVβ3 dependent manner in vitro. Inhibiting S1PR2 signaling with specific inhibitor JTE-013 or siRNA decreased Mfge8 expression and impaired the erythrophagocytosis of VSMCs in vitro. Partial ligation was performed in the left common carotid artery (LCA) followed by intra-intimal injection of isolated erythrocytes to observe their clearance in vivo. Interfering S1PR2 expression in VSMCs with Adeno-associated virus 9 inhibited MFG-E8 expression inside LCA plaques receiving RBCs injection and attenuated erythrocytes clearance. Erythrophagocytosis by VSMCs increased vascular endothelial growth factor-a secretion and promoted angiogenesis. The present study revealed that VSMCs act as phagocytes for RBC clearance through S1PR2 activation induced MFG-E8 release.
Collapse
Affiliation(s)
- Daorong Pan
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Wen Wu
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Guangfeng Zuo
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Xiangrong Xie
- Department of Cardiology, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu 241001, Anhui, China
| | - Hui Li
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Xiaomin Ren
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Chaohua Kong
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Wenying Zhou
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Zihan Zhang
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Martin Waterfall
- Institute of Immunology & Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH8 9JZ, United Kingdom
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital, The Affiliated Nanjing Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210006, Jiangsu, China.
| |
Collapse
|
8
|
Cabrera JTO, Makino A. Efferocytosis of vascular cells in cardiovascular disease. Pharmacol Ther 2022; 229:107919. [PMID: 34171333 PMCID: PMC8695637 DOI: 10.1016/j.pharmthera.2021.107919] [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: 01/25/2021] [Revised: 05/21/2021] [Accepted: 06/03/2021] [Indexed: 12/20/2022]
Abstract
Cell death and the clearance of apoptotic cells are tightly regulated by various signaling molecules in order to maintain physiological tissue function and homeostasis. The phagocytic removal of apoptotic cells is known as the process of efferocytosis, and abnormal efferocytosis is linked to various health complications and diseases, such as cardiovascular disease, inflammatory diseases, and autoimmune diseases. During efferocytosis, phagocytic cells and/or apoptotic cells release signals, such as "find me" and "eat me" signals, to stimulate the phagocytic engulfment of apoptotic cells. Primary phagocytic cells are macrophages and dendritic cells; however, more recently, other neighboring cell types have also been shown to exhibit phagocytic character, including endothelial cells and fibroblasts, although they are comparatively slower in clearing dead cells. In this review, we focus on macrophage efferocytosis of vascular cells, such as endothelial cells, smooth muscle cells, fibroblasts, and pericytes, and its relation to the progression and development of cardiovascular disease. We also highlight the role of efferocytosis-related molecules and their contribution to the maintenance of vascular homeostasis.
Collapse
Affiliation(s)
- Jody Tori O Cabrera
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
9
|
Ji Y, Lisabeth EM, Neubig RR. Transforming Growth Factor β1 Increases Expression of Contractile Genes in Human Pulmonary Arterial Smooth Muscle Cells by Potentiating Sphingosine-1-Phosphate Signaling. Mol Pharmacol 2021; 100:53-60. [PMID: 34031187 DOI: 10.1124/molpharm.120.000019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/30/2021] [Indexed: 11/22/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by elevated pulmonary arterial pressure and carries a very poor prognosis. Understanding of PAH pathogenesis is needed to support the development of new therapeutic strategies. Transforming growth factor β (TGF-β) drives vascular remodeling and increases vascular resistance by regulating differentiation and proliferation of smooth muscle cells (SMCs). Also, sphingosine-1-phosphate (S1P) has been implicated in PAH, but the relation between these two signaling mechanisms is not well understood. Here, we characterize the signaling networks downstream of TGF-β in human pulmonary arterial smooth muscle cells (HPASMCs), which involves mothers against decapentaplegic homolog (SMAD) signaling as well as Rho GTPases. Activation of Rho GTPases regulates myocardin-related transcription factor (MRTF) and serum response factor (SRF) transcription activity and results in upregulation of contractile gene expression. Our genetic and pharmacologic data show that in HPASMCs upregulation of α smooth muscle actin (αSMA) and calponin by TGF-β is dependent on both SMAD and Rho/MRTF-A/SRF transcriptional mechanisms.The kinetics of TGF-β-induced myosin light chain (MLC) 2 phosphorylation, a measure of RhoA activation, are slow, as is regulation of the Rho/MRTF/SRF-induced αSMA expression. These results suggest that TGF-β1 activates Rho/phosphorylated MLC2 through an indirect mechanism, which was confirmed by sensitivity to cycloheximide treatment. As a potential mechanism for this indirect action, TGF-β1 upregulates mRNA for sphingosine kinase (SphK1), the enzyme that produces S1P, an upstream Rho activator, as well as mRNA levels of the S1P receptor (S1PR) 3. SphK1 inhibitor and S1PR3 inhibitors (PF543 and TY52156/VPC23019) reduce TGF-β1-induced αSMA upregulation. Overall, we propose a model in which TGF-β1 activates Rho/MRTF-A/SRF by potentiating an autocrine/paracrine S1P signaling mechanism through SphK1 and S1PR3. SIGNIFICANCE STATEMENT: In human pulmonary arterial smooth muscle cells, transforming growth factor β depends on sphingosine-1-phosphate signaling to bridge the interaction between mothers against decapentaplegic homolog and Rho/myocardin-related transcription factor (MRTF) signaling in regulating α smooth muscle actin (αSMA) expression. The Rho/MRTF pathway is a signaling node in the αSMA regulatory network and is a potential therapeutic target for the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Yajing Ji
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (Y.J., E.M.L., R.R.N.) and Nicholas V. Perricone, MD, Division of Dermatology, Department of Medicine, College of Human Medicine, East Lansing, Michigan (R.R.N.)
| | - Erika M Lisabeth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (Y.J., E.M.L., R.R.N.) and Nicholas V. Perricone, MD, Division of Dermatology, Department of Medicine, College of Human Medicine, East Lansing, Michigan (R.R.N.)
| | - Richard R Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan (Y.J., E.M.L., R.R.N.) and Nicholas V. Perricone, MD, Division of Dermatology, Department of Medicine, College of Human Medicine, East Lansing, Michigan (R.R.N.)
| |
Collapse
|
10
|
Sarver DC, Lei X, Wong GW. FAM19A (TAFA): An Emerging Family of Neurokines with Diverse Functions in the Central and Peripheral Nervous System. ACS Chem Neurosci 2021; 12:945-958. [PMID: 33621067 DOI: 10.1021/acschemneuro.0c00757] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cytokines and chemokines have diverse and pleiotropic functions in peripheral tissues and in the brain. Recent studies uncovered a novel family of neuron-derived secretory proteins, or neurokines, distantly related to chemokines. The FAM19A family comprises five ∼12-15 kDa secretory proteins (FAM19A1-5), also known as TAFA1-5, that are predominantly detected in the central and peripheral nervous system. FAM19A expression in the central nervous system is dynamically regulated during development and in the postnatal brain. As secreted ligands, FAM19A proteins appear to bind to different classes of cell surface receptors (e.g., GPCRs and neurexins). Functional studies using gain- and loss-of-function mouse models established nonredundant roles for each FAM19A family member in regulating diverse physiological processes ranging from locomotor activity and food intake to learning and memory, anxiety- and depressive-like behaviors, social communication, repetitive behaviors, and somatosensory functions. This review summarizes major advances as well as the limitations and knowledge gaps in understanding the regulation and diverse biological functions of this conserved family of neurokines.
Collapse
Affiliation(s)
- Dylan C. Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Xia Lei
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - G. William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
11
|
Li Y, Li Y, Jing X, Liu Y, Liu B, She Q. Sphingosine 1-phosphate induces epicardial progenitor cell differentiation into smooth muscle-like cells. Acta Biochim Biophys Sin (Shanghai) 2019; 51:402-410. [PMID: 30877755 DOI: 10.1093/abbs/gmz017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 01/29/2019] [Accepted: 02/01/2019] [Indexed: 11/13/2022] Open
Abstract
Epicardial progenitor cells (EpiCs) which are derived from the proepicardium have the potential to differentiate into coronary vascular smooth muscle cells during development. Whether sphingosine 1-phosphate (S1P), a highly hydrophobic zwitterionic lysophospholipid in signal transduction, induces the differentiation of EpiCs is unknown. In the present study, we demonstrated that S1P significantly induced the expression of smooth muscle cell specific markers α-smooth muscle actin and myosin heavy chain 11 in the EpiCs. And the smooth muscle cells differentiated from the EpiCs stimulated by S1P were further evaluated by gel contraction assay. To further confirm the major subtype of sphingosine 1-phosphate receptors (S1PRs) involved in the differentiation of EpiCs, we used the agonists and antagonists of different S1PRs. The results showed that the S1P1/S1P3 antagonist VPC23019 and the S1P2 antagonist JTE013 significantly attenuated EpiCs differentiation, while the S1P1 agonist SEW2871 and antagonist W146 did not affect EpiCs differentiation. These results collectively suggested that S1P, principally through its receptor S1P3, increases EpiCs differentiation into VSMCs and thus indicated the importance of S1P signaling in the embryonic coronary vasculature, while S1P2 plays a secondary role.
Collapse
Affiliation(s)
- Yu Li
- Department of Cardiology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yingrui Li
- Department of Cardiology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xiaodong Jing
- Department of Cardiology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yajie Liu
- Department of Cardiology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Bin Liu
- Department of Cardiology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qiang She
- Department of Cardiology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Kitano T, Usui S, Takashima SI, Inoue O, Goten C, Nomura A, Yoshioka K, Okajima M, Kaneko S, Takuwa Y, Takamura M. Sphigosine-1-phosphate receptor 1 promotes neointimal hyperplasia in a mouse model of carotid artery injury. Biochem Biophys Res Commun 2019; 511:179-184. [DOI: 10.1016/j.bbrc.2019.02.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/08/2019] [Indexed: 11/28/2022]
|
13
|
Jin J, Lu Z, Li Y, Ru JH, Lopes-Virella MF, Huang Y. LPS and palmitate synergistically stimulate sphingosine kinase 1 and increase sphingosine 1 phosphate in RAW264.7 macrophages. J Leukoc Biol 2018; 104:843-853. [PMID: 29882996 PMCID: PMC6162112 DOI: 10.1002/jlb.3a0517-188rrr] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 05/15/2018] [Accepted: 05/18/2018] [Indexed: 01/28/2023] Open
Abstract
It has been well established that patients with diabetes or metabolic syndrome (MetS) have increased prevalence and severity of periodontitis, an oral infection initiated by bacteria and characterized by tissue inflammation and destruction. To understand the underlying mechanisms, we have shown that saturated fatty acid (SFA), which is increased in patients with type 2 diabetes or MetS, and LPS, an important pathogenic factor for periodontitis, synergistically stimulate expression of proinflammatory cytokines in macrophages by increasing ceramide production. However, the mechanisms by which increased ceramide enhances proinflammatory cytokine expression have not been well understood. Since sphingosine 1 phosphate (S1P) is a metabolite of ceramide and a bioactive lipid, we tested our hypothesis that stimulation of ceramide production by LPS and SFA facilitates S1P production, which contributes to proinflammatory cytokine expression. Results showed that LPS and palmitate, a major SFA, synergistically increased not only ceramide, but also S1P, and stimulated sphingosine kinase (SK) expression and membrane translocation in RAW264.7 macrophages. Results also showed that SK inhibition attenuated the stimulatory effect of LPS and palmitate on IL-6 secretion. Moreover, results showed that S1P enhanced the stimulatory effect of LPS and palmitate on IL-6 secretion. Finally, results showed that targeting S1P receptors using either S1P receptor antagonists or small interfering RNA attenuated IL-6 upregulation by LPS and palmitate. Taken together, this study demonstrated that LPS and palmitate synergistically stimulated S1P production and S1P in turn contributed to the upregulation of proinflammatory cytokine expression in macrophages by LPS and palmitate.
Collapse
Affiliation(s)
- Junfei Jin
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Zhongyang Lu
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA
| | - Yanchun Li
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ji Hyun Ru
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Maria F Lopes-Virella
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA
| | - Yan Huang
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
14
|
Jadczyk T, Baranski K, Syzdol M, Nabialek E, Wanha W, Kurzelowski R, Ratajczak MZ, Kucia M, Dolegowska B, Niewczas M, Zejda J, Wojakowski W. Bioactive Sphingolipids, Complement Cascade, and Free Hemoglobin Levels in Stable Coronary Artery Disease and Acute Myocardial Infarction. Mediators Inflamm 2018; 2018:2691934. [PMID: 30116144 PMCID: PMC6079520 DOI: 10.1155/2018/2691934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/29/2018] [Accepted: 03/13/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) and coronary artery bypass graft (CABG) surgery are associated with a pathogen-free inflammatory response (sterile inflammation). Complement cascade (CC) and bioactive sphingolipids (BS) are postulated to be involved in this process. AIM The aim of this study was to evaluate plasma levels of CC cleavage fragments (C3a, C5a, and C5b9), sphingosine (SP), sphingosine-1-phosphate (S1P), and free hemoglobin (fHb) in AMI patients treated with primary percutaneous coronary intervention (pPCI) and stable coronary artery disease (SCAD) undergoing CABG. PATIENTS AND METHODS The study enrolled 37 subjects (27 male) including 22 AMI patients, 7 CABG patients, and 8 healthy individuals as the control group (CTRL). In the AMI group, blood samples were collected at 5 time points (admission to hospital, 6, 12, 24, and 48 hours post pPCI) and 4 time points in the CABG group (6, 12, 24, and 48 hours post operation). SP and S1P concentrations were measured by high-performance liquid chromatography (HPLC). Analysis of C3a, C5a, and C5b9 levels was carried out using high-sensitivity ELISA and free hemoglobin by spectrophotometry. RESULTS The plasma levels of CC cleavage fragments (C3a and C5b9) were significantly higher, while those of SP and S1P were lower in patients undergoing CABG surgery in comparison to the AMI group. In both groups, levels of CC factors showed no significant changes within 48 hours of follow-up. Conversely, SP and S1P levels gradually decreased throughout 48 hours in the AMI group but remained stable after CABG. Moreover, the fHb concentration was significantly higher after 24 and 48 hours post pPCI compared to the corresponding postoperative time points. Additionally, the fHb concentrations increased between 12 and 48 hours after PCI in patients with AMI. CONCLUSIONS Inflammatory response after AMI and CABG differed regarding the release of sphingolipids, free hemoglobin, and complement cascade cleavage fragments.
Collapse
Affiliation(s)
- T. Jadczyk
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Ziołowa 45-47, Katowice, Poland
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - K. Baranski
- Department of Epidemiology, Medical University of Silesia, Katowice, Poland
| | - M. Syzdol
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Ziołowa 45-47, Katowice, Poland
| | - E. Nabialek
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Ziołowa 45-47, Katowice, Poland
| | - W. Wanha
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Ziołowa 45-47, Katowice, Poland
| | - R. Kurzelowski
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Ziołowa 45-47, Katowice, Poland
| | - M. Z. Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - M. Kucia
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - B. Dolegowska
- Department of Laboratory Medicine, Pomeranian Medical University, Szczecin, Poland
| | - M. Niewczas
- Department of Sport, Faculty of Physical Education, University of Rzeszow, Rzeszow, Poland
| | - J. Zejda
- Department of Epidemiology, Medical University of Silesia, Katowice, Poland
| | - W. Wojakowski
- Division of Cardiology and Structural Heart Diseases, Medical University of Silesia, Ziołowa 45-47, Katowice, Poland
| |
Collapse
|
15
|
Wang Y, Chen D, Zhang Y, Wang P, Zheng C, Zhang S, Yu B, Zhang L, Zhao G, Ma B, Cai Z, Xie N, Huang S, Liu Z, Mo X, Guan Y, Wang X, Fu Y, Ma D, Wang Y, Kong W. Novel Adipokine, FAM19A5, Inhibits Neointima Formation After Injury Through Sphingosine-1-Phosphate Receptor 2. Circulation 2018; 138:48-63. [DOI: 10.1161/circulationaha.117.032398] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 02/01/2018] [Indexed: 01/10/2023]
Abstract
Background:
Obesity plays crucial roles in the development of cardiovascular diseases. However, the mechanisms that link obesity and cardiovascular diseases remain elusive. Compelling evidence indicates that adipokines play an important role in obesity-related cardiovascular diseases. Here, we found a new adipokine-named family with sequence similarity 19, member A5 (FAM19A5), a protein with unknown function that was predicted to be distantly related to the CC-chemokine family. We aimed to test whether adipose-derived FAM19A5 regulates vascular pathology on injury.
Methods:
DNA cloning, protein expression, purification, and N-terminal sequencing were applied to characterize FAM19A5. Adenovirus infection and siRNA transfection were performed to regulate FAM19A5 expression. Balloon and wire injury were performed in vivo on the rat carotid arteries and mouse femoral arteries, respectively. Bioinformatics analysis, radioactive ligand-receptor binding assays, receptor internalization, and calcium mobilization assays were used to identify the functional receptor for FAM19A5.
Results:
We first characterized FAM19A5 as a secreted protein, and the first 43 N-terminal amino acids were the signal peptides. Both FAM19A5 mRNA and protein were abundantly expressed in the adipose tissue but were downregulated in obese mice. Overexpression of FAM19A5 markedly inhibited vascular smooth muscle cell proliferation and migration and neointima formation in the carotid arteries of balloon-injured rats. Accordingly, FAM19A5 silencing in adipocytes significantly promoted vascular smooth muscle cell activation. Adipose-specific FAM19A5 transgenic mice showed greater attenuation of neointima formation compared with wild-type littermates fed with or without Western-style diet. We further revealed that sphingosine-1-phosphate receptor 2 was the functional receptor for FAM19A5, with a dissociation constant (
K
d
) of 0.634 nmol/L. Inhibition of sphingosine-1-phosphate receptor 2 or its downstream G12/13-RhoA signaling circumvented the suppressive effects of FAM19A5 on vascular smooth muscle cell proliferation and migration.
Conclusions:
We revealed that a novel adipokine, FAM19A5, was capable of inhibiting postinjury neointima formation via sphingosine-1-phosphate receptor 2-G12/13-RhoA signaling. Downregulation of FAM19A5 during obesity may trigger cardiometabolic diseases.
Collapse
Affiliation(s)
- Yingbao Wang
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Dixin Chen
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yan Zhang
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Pingzhang Wang
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China (P.W., X.M., D.M., Y.W.)
| | - Can Zheng
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Songyang Zhang
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Bing Yu
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Lu Zhang
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Guizhen Zhao
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Baihui Ma
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Zeyu Cai
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Nan Xie
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Shiyang Huang
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ziyi Liu
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Xiaoning Mo
- Center for Human Disease Genomics, Peking University, Beijing, China (P.W., X.M., D.M., Y.W.)
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Liaoning, China (Y.G.)
| | - Xian Wang
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Yi Fu
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| | - Dalong Ma
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China (P.W., X.M., D.M., Y.W.)
| | - Ying Wang
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health (D.C., Y.Z., P.W., C.Z., S.H., D.M., Y.W.), School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Center for Human Disease Genomics, Peking University, Beijing, China (P.W., X.M., D.M., Y.W.)
| | - Wei Kong
- Department of Physiology and Pathophysiology (Y.W., D.C., S.Z., B.Y., L.Z., G.Z., B.M., Z.C., N.X., Z.L., X.W., Y.F., W.K.)
| |
Collapse
|
16
|
Braetz J, Becker A, Geissen M, Larena-Avellaneda A, Schrepfer S, Daum G. Sphingosine-1-phosphate receptor 1 regulates neointimal growth in a humanized model for restenosis. J Vasc Surg 2018; 68:201S-207S. [PMID: 29804740 DOI: 10.1016/j.jvs.2018.02.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/28/2018] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The main objective of this study was to define a role of sphingosine-1-phosphate receptor 1 (S1PR1) in the arterial injury response of a human artery. The hypotheses were tested that injury induces an expansion of S1PR1-positive cells and that these cells accumulate toward the lumen because they follow the sphingosine-1-phosphate gradient from arterial wall tissue (low) to plasma (high). METHODS A humanized rat model was used in which denuded human internal mammary artery (IMA) was implanted into the position of the abdominal aorta of immunosuppressed Rowett nude rats. This injury model is characterized by medial as well as intimal hyperplasia, whereby intimal cells are of human origin. At 7, 14, and 28 days after implantation, grafts were harvested and processed for fluorescent immunostaining for S1PR1 and smooth muscle α-actin. Nuclei were stained with 4',6-diamidine-2'-phenylindole dihydrochloride. Using digitally reconstructed, complete cross sections of grafts, intimal and medial areas were measured, whereby the medial area had virtually been divided into an outer (toward adventitia) and inner (toward lumen) layer. The fraction of S1PR1-positive cells was determined in each layer by counting S1PR1-positive and S1PR1-negative cells. RESULTS The fraction of S1PR1-postive cells in naive IMA is 58.9% ± 6.0% (mean ± standard deviation). At day 28 after implantation, 81.6% ± 4.4% of medial cells were scored S1PR1 positive (P < .01). At day 14, the ratio between S1PR1-positive and S1PR1-negative cells was significantly higher in the lumen-oriented inner layer (9.3 ± 2.1 vs 6.0 ± 1.0; P < .01). Cells appearing in the intima at day 7 and day 14 were almost all S1PR1 positive. At day 28, however, about one-third of intimal cells were scored S1PR1 negative. CONCLUSIONS From these data, we conclude that denudation of IMA specifically induces the expansion of S1PR1-positive cells. Based on the nonrandom distribution of S1PR1-positive cells, we consider the possibility that much like lymphocytes, S1PR1-positive smooth muscle cells also use S1PR1 to recognize the sphingosine-1-phosphate gradient from tissue (low) to plasma (high) and so migrate out of the media toward the intima of the injured IMA.
Collapse
Affiliation(s)
- Julian Braetz
- Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, Hamburg, Germany; Clinic and Polyclinic for General and Interventional Cardiology, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Astrid Becker
- Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Geissen
- Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Axel Larena-Avellaneda
- Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonja Schrepfer
- Clinic and Polyclinic for Cardiovascular Surgery, Transplant and Stem Cell Immunobiology Laboratory, University Heart Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guenter Daum
- Clinic and Polyclinic for Vascular Medicine, University Heart Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
17
|
Liu H, Jin H, Yue X, Han J, Baum P, Abendschein DR, Tu Z. PET Study of Sphingosine-1-Phosphate Receptor 1 Expression in Response to Vascular Inflammation in a Rat Model of Carotid Injury. Mol Imaging 2018; 16:1536012116689770. [PMID: 28654378 PMCID: PMC5470136 DOI: 10.1177/1536012116689770] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Sphingosine-1-phosphate receptor (S1PR) activation plays a key role in vascular inflammatory response. Here, we report in vivo validation of [11C]TZ3321, a potent S1PR1 radioligand, for imaging vascular inflammation in a rat model of carotid injury. The right common carotid artery of male adult Sprague-Dawley rats was injured by balloon overinflation that denuded the endothelium and distended the vessel wall. Animals received a 60-minute micro-positron emission tomography (micro PET) scan with [11C]TZ3321 at 72 hours after injury. Ex vivo autoradiography was also conducted. The expression and cellular location of S1PR1 were examined by immunohistological analysis. Three-dimensional (3D) reconstruction of the first 100-second microPET/computed tomography (CT) image indicated the location of bilateral common carotid arteries. [11C]TZ3321 displayed significantly higher accumulation (standardized uptake values: 0.93 ± 0.07 vs 0.78 ± 0.09, n = 6, P = .001) in the injured carotid artery than in the contralateral side. Increased tracer uptake in the injured artery was confirmed by autoradiography (photostimulated luminescence measures: 85.5 ± 0.93 vs 71.48 ± 6.22, n = 2). Concordantly, high S1PR1expression was observed in infiltrated inflammatory cells in the injured artery. Our studies demonstrate [11C]TZ3321 microPET is able to detect the acute upregulation of S1PR1 expression in inflamed carotid artery. Therefore, [11C]TZ3321 has potential to be a PET radiotracer for detecting early inflammatory response and monitoring therapeutic efficacy of vascular inflammation.
Collapse
Affiliation(s)
- Hui Liu
- 1 Department of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Hongjun Jin
- 1 Department of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Xuyi Yue
- 1 Department of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Junbin Han
- 1 Department of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Pamela Baum
- 2 Center for Cardiovascular Research, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Dana R Abendschein
- 2 Center for Cardiovascular Research, Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Zhude Tu
- 1 Department of Radiology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
18
|
Lee KP, Baek S, Jung SH, Cui L, Lee D, Lee DY, Choi WS, Chung HW, Lee BH, Kim B, Won KJ. DJ-1 is involved in epigenetic control of sphingosine-1-phosphate receptor expression in vascular neointima formation. Pflugers Arch 2018; 470:1103-1113. [DOI: 10.1007/s00424-018-2132-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/12/2018] [Accepted: 02/21/2018] [Indexed: 01/30/2023]
|
19
|
Vestri A, Pierucci F, Frati A, Monaco L, Meacci E. Sphingosine 1-Phosphate Receptors: Do They Have a Therapeutic Potential in Cardiac Fibrosis? Front Pharmacol 2017. [PMID: 28626422 PMCID: PMC5454082 DOI: 10.3389/fphar.2017.00296] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid that is characterized by a peculiar mechanism of action. In fact, S1P, which is produced inside the cell, can act as an intracellular mediator, whereas after its export outside the cell, it can act as ligand of specific G-protein coupled receptors, which were initially named endothelial differentiation gene (Edg) and eventually renamed sphingosine 1-phosphate receptors (S1PRs). Among the five S1PR subtypes, S1PR1, S1PR2 and S1PR3 isoforms show broad tissue gene expression, while S1PR4 is primarily expressed in immune system cells, and S1PR5 is expressed in the central nervous system. There is accumulating evidence for the important role of S1P as a mediator of many processes, such as angiogenesis, carcinogenesis and immunity, and, ultimately, fibrosis. After a tissue injury, the imbalance between the production of extracellular matrix (ECM) and its degradation, which occurs due to chronic inflammatory conditions, leads to an accumulation of ECM and, consequential, organ dysfunction. In these pathological conditions, many factors have been described to act as pro- and anti-fibrotic agents, including S1P. This bioactive lipid exhibits both pro- and anti-fibrotic effects, depending on its site of action. In this review, after a brief description of sphingolipid metabolism and signaling, we emphasize the involvement of the S1P/S1PR axis and the downstream signaling pathways in the development of fibrosis. The current knowledge of the therapeutic potential of S1PR subtype modulators in the treatment of the cardiac functions and fibrinogenesis are also examined.
Collapse
Affiliation(s)
- Ambra Vestri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy
| | - Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy.,Interuniversity Institutes of MyologyFirenze, Italy
| | - Alessia Frati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy
| | - Lucia Monaco
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of RomeRome, Italy
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Molecular and Applied Biology Research Unit, University of FlorenceFlorence, Italy.,Interuniversity Institutes of MyologyFirenze, Italy
| |
Collapse
|
20
|
Thackeray JT, Bengel FM. Specificity vs versatility: A fine balance for novel targeted molecular imaging radiotracers. J Nucl Cardiol 2017; 24:571-573. [PMID: 26864090 DOI: 10.1007/s12350-016-0426-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 01/21/2016] [Indexed: 12/23/2022]
Affiliation(s)
- James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
21
|
Jin H, Yang H, Liu H, Zhang Y, Zhang X, Rosenberg AJ, Liu Y, Lapi SE, Tu Z. A promising carbon-11-labeled sphingosine-1-phosphate receptor 1-specific PET tracer for imaging vascular injury. J Nucl Cardiol 2017; 24:558-570. [PMID: 26843200 DOI: 10.1007/s12350-015-0391-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 12/04/2015] [Indexed: 01/19/2023]
Abstract
BACKGROUND Sphingosine-1-phosphate receptor 1 (S1PR1) is highly expressed in vascular smooth muscle cells from intimal lesions. PET imaging using S1PR1 as a biomarker would increase our understanding of its role in vascular pathologies including in-stent restenosis. METHODS The S1PR1 compound TZ3321 was synthesized for in vitro characterization and labeled with Carbon-11 for in vivo studies. The biodistribution of [11C]TZ3321 was evaluated in normal mice; microPET and immunohistochemistry (IHC) studies were performed using a murine femoral artery wire-injury model of restenosis. RESULTS The high potency of TZ3321 for S1PR1 (IC 50 = 2.13 ± 1.63 nM), and high selectivity (>1000 nM) for S1PR1 over S1PR2 and S1PR3 were confirmed. Biodistribution data revealed prolonged retention of [11C]TZ3321 in S1PR1-enriched tissues. MicroPET imaging of [11C]TZ3321 showed higher uptake in the wire-injured arteries of ApoE-/- mice than in injured arteries of wild-type mice (SUV 0.40 ± 0.06 vs 0.28 ± 0.04, n = 6, P < .001); FDG-PET showed no difference (SUV 0.98 ± 0.04 vs 0.94 ± 0.01, n = 6, P > .05). Post-PET autoradiography showed >4-fold higher [11C]TZ3321 retention in the injured artery of ApoE-/- mice than in wild-type mice. Subsequent IHC staining confirmed higher expression of S1PR1 in the neointima of the injured artery of ApoE-/- mice than in wild-type mice. CONCLUSIONS This preliminary study supports the potential use of PET for quantification of the S1PR1 expression as a biomarker of neointimal hyperplasia.
Collapse
Affiliation(s)
- Hongjun Jin
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Boulevard, St. Louis, MO, 63110, USA
| | - Hao Yang
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Boulevard, St. Louis, MO, 63110, USA
| | - Hui Liu
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Boulevard, St. Louis, MO, 63110, USA
| | - Yunxiao Zhang
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Boulevard, St. Louis, MO, 63110, USA
| | - Xiang Zhang
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Boulevard, St. Louis, MO, 63110, USA
| | - Adam J Rosenberg
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Boulevard, St. Louis, MO, 63110, USA
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Boulevard, St. Louis, MO, 63110, USA
| | - Suzanne E Lapi
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Boulevard, St. Louis, MO, 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Boulevard, St. Louis, MO, 63110, USA.
| |
Collapse
|
22
|
Christoffersen C, Bartels ED, Aarup A, Nielsen LB, Pedersen TX. ApoB and apoM - New aspects of lipoprotein biology in uremia-induced atherosclerosis. Eur J Pharmacol 2017; 816:154-160. [PMID: 28351665 DOI: 10.1016/j.ejphar.2017.03.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/09/2017] [Accepted: 03/23/2017] [Indexed: 12/27/2022]
Abstract
Chronic kidney disease affects as much as 13% of the population, and is associated with a markedly increased risk of developing cardiovascular disease. One of the underlying reasons is accelerated development of atherosclerosis. This can be ascribed both to increased occurrence of traditional cardiovascular risk factors, and to risk factors that may be unique to patients with chronic kidney disease. The latter is reflected in the observation that the current treatment modalities, mainly directed against traditional risk factors, are insufficient to prevent cardiovascular disease in the patient with chronic kidney disease. This review discusses mechanisms accelerating uremic atherosclerosis with a specific focus on the putative roles of apolipoprotein(apo)s B and M that may be particularly important in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Oe, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Emil D Bartels
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Oe, Denmark.
| | - Annemarie Aarup
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Lars B Nielsen
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen Oe, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark; Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Tanja X Pedersen
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| |
Collapse
|
23
|
Riascos-Bernal DF, Chinnasamy P, Gross JN, Almonte V, Egaña-Gorroño L, Parikh D, Jayakumar S, Guo L, Sibinga NES. Inhibition of Smooth Muscle β-Catenin Hinders Neointima Formation After Vascular Injury. Arterioscler Thromb Vasc Biol 2017; 37:879-888. [PMID: 28302627 DOI: 10.1161/atvbaha.116.308643] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 03/01/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Smooth muscle cells (SMCs) contribute to neointima formation after vascular injury. Although β-catenin expression is induced after injury, whether its function is essential in SMCs for neointimal growth is unknown. Moreover, although inhibitors of β-catenin have been developed, their effects on SMC growth have not been tested. We assessed the requirement for SMC β-catenin in short-term vascular homeostasis and in response to arterial injury and investigated the effects of β-catenin inhibitors on vascular SMC growth. APPROACH AND RESULTS We used an inducible, conditional genetic deletion of β-catenin in SMCs of adult mice. Uninjured arteries from adult mice lacking SMC β-catenin were indistinguishable from controls in terms of structure and SMC marker gene expression. After carotid artery ligation, however, vessels from mice lacking SMC β-catenin developed smaller neointimas, with lower neointimal cell proliferation and increased apoptosis. SMCs lacking β-catenin showed decreased mRNA expression of Mmp2, Mmp9, Sphk1, and S1pr1 (genes that promote neointima formation), higher levels of Jag1 and Gja1 (genes that inhibit neointima formation), decreased Mmp2 protein expression and secretion, and reduced cell invasion in vitro. Moreover, β-catenin inhibitors PKF118-310 and ICG-001 limited growth of mouse and human vascular SMCs in a dose-dependent manner. CONCLUSIONS SMC β-catenin is dispensable for maintenance of the structure and state of differentiation of uninjured adult arteries, but is required for neointima formation after vascular injury. Pharmacological β-catenin inhibitors hinder growth of human vascular SMCs. Thus, inhibiting β-catenin has potential as a therapy to limit SMC accumulation and vascular obstruction.
Collapse
Affiliation(s)
- Dario F Riascos-Bernal
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (D.F.R.-B., P.C., J.N.G., V.A., L.E.-G., D.P., S.J., N.E.S.S.); and CVPath Institute, Gaithersburg, MD (L.G.)
| | - Prameladevi Chinnasamy
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (D.F.R.-B., P.C., J.N.G., V.A., L.E.-G., D.P., S.J., N.E.S.S.); and CVPath Institute, Gaithersburg, MD (L.G.)
| | - Jordana N Gross
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (D.F.R.-B., P.C., J.N.G., V.A., L.E.-G., D.P., S.J., N.E.S.S.); and CVPath Institute, Gaithersburg, MD (L.G.)
| | - Vanessa Almonte
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (D.F.R.-B., P.C., J.N.G., V.A., L.E.-G., D.P., S.J., N.E.S.S.); and CVPath Institute, Gaithersburg, MD (L.G.)
| | - Lander Egaña-Gorroño
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (D.F.R.-B., P.C., J.N.G., V.A., L.E.-G., D.P., S.J., N.E.S.S.); and CVPath Institute, Gaithersburg, MD (L.G.)
| | - Dippal Parikh
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (D.F.R.-B., P.C., J.N.G., V.A., L.E.-G., D.P., S.J., N.E.S.S.); and CVPath Institute, Gaithersburg, MD (L.G.)
| | - Smitha Jayakumar
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (D.F.R.-B., P.C., J.N.G., V.A., L.E.-G., D.P., S.J., N.E.S.S.); and CVPath Institute, Gaithersburg, MD (L.G.)
| | - Liang Guo
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (D.F.R.-B., P.C., J.N.G., V.A., L.E.-G., D.P., S.J., N.E.S.S.); and CVPath Institute, Gaithersburg, MD (L.G.)
| | - Nicholas E S Sibinga
- From the Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY (D.F.R.-B., P.C., J.N.G., V.A., L.E.-G., D.P., S.J., N.E.S.S.); and CVPath Institute, Gaithersburg, MD (L.G.).
| |
Collapse
|
24
|
Kanemura N, Shibata R, Ohashi K, Ogawa H, Hiramatsu-Ito M, Enomoto T, Yuasa D, Ito M, Hayakawa S, Otaka N, Murohara T, Ouchi N. C1q/TNF-related protein 1 prevents neointimal formation after arterial injury. Atherosclerosis 2017; 257:138-145. [PMID: 28131048 DOI: 10.1016/j.atherosclerosis.2017.01.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 12/08/2016] [Accepted: 01/13/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIMS Obesity contributes to the progression of vascular disorders. C1q/TNF-related protein (CTRP) 1 is a circulating adipokine, which is upregulated in obese complications including coronary artery disease. Here, we investigated the role of CTRP1 in regulation of vascular remodeling after mechanical injury and evaluated its potential mechanism. METHODS Mice were subjected to wire-induced injury of left femoral arteries. An adenoviral vector encoding CTRP1 (Ad-CTRP1) or β-galactosidase as a control was injected into the jugular vein of mice 3 days prior to surgery. RESULTS Systemic administration of Ad-CTRP1 to wild-type mice led to reduction of the neointimal thickening after wire-induced arterial injury and the number of bromodeoxyuridine-positive cells in injured vessels as compared with treatment with control vectors. Treatment of vascular smooth muscle cells (VSMCs) with CTRP1 protein attenuated proliferative activity and ERK phosphorylation in response to PDGF-BB. CTRP1 treatment increased cyclic AMP (cAMP) levels in VSMCs, and inhibition of adenylyl cyclase reversed the inhibitory effect of CTRP1 on VSMC growth and ERK phosphorylation. Antagonization of sphingosine-1-phosphaterote (S1P) receptor 2 blocked the effects of CTRP1 on cAMP production and VSMC growth. Furthermore, CTRP1-knockout mice had enhanced neointimal thickening following injury and increased numbers of proliferating cells in neointima compared to control WT mice. CONCLUSIONS These findings indicate that CTRP1 functions to prevent the development of pathological vascular remodeling by reducing VSMC growth through the cAMP-dependent pathway.
Collapse
MESH Headings
- Adipokines/deficiency
- Adipokines/genetics
- Adipokines/metabolism
- Animals
- Cell Proliferation
- Cells, Cultured
- Cyclic AMP/metabolism
- Disease Models, Animal
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Femoral Artery/injuries
- Femoral Artery/metabolism
- Femoral Artery/pathology
- Genetic Predisposition to Disease
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phenotype
- Phosphorylation
- Proteins/metabolism
- Receptors, Lysosphingolipid/metabolism
- Signal Transduction
- Vascular System Injuries/genetics
- Vascular System Injuries/metabolism
- Vascular System Injuries/pathology
Collapse
Affiliation(s)
- Noriyoshi Kanemura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rei Shibata
- Department of Advanced Cardiovascular Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Koji Ohashi
- Molecular Cardiovascular Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hayato Ogawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mizuho Hiramatsu-Ito
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Enomoto
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Yuasa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masanori Ito
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoko Hayakawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoya Otaka
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noriyuki Ouchi
- Molecular Cardiovascular Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
25
|
Chew WS, Wang W, Herr DR. To fingolimod and beyond: The rich pipeline of drug candidates that target S1P signaling. Pharmacol Res 2016; 113:521-532. [DOI: 10.1016/j.phrs.2016.09.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/20/2016] [Accepted: 09/20/2016] [Indexed: 01/28/2023]
|
26
|
Blankenbach KV, Schwalm S, Pfeilschifter J, Meyer Zu Heringdorf D. Sphingosine-1-Phosphate Receptor-2 Antagonists: Therapeutic Potential and Potential Risks. Front Pharmacol 2016; 7:167. [PMID: 27445808 PMCID: PMC4914510 DOI: 10.3389/fphar.2016.00167] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/03/2016] [Indexed: 12/26/2022] Open
Abstract
The sphingosine-1-phosphate (S1P) signaling system with its specific G-protein-coupled S1P receptors, the enzymes of S1P metabolism and the S1P transporters, offers a multitude of promising targets for drug development. Until today, drug development in this area has nearly exclusively focused on (functional) antagonists at the S1P1 receptor, which cause a unique phenotype of immunomodulation. Accordingly, the first-in class S1P1 receptor modulator, fingolimod, has been approved for the treatment of relapsing-remitting multiple sclerosis, and novel S1P1 receptor (functional) antagonists are being developed for autoimmune and inflammatory diseases such as psoriasis, inflammatory bowel disease, lupus erythematodes, or polymyositis. Besides the S1P1 receptor, also S1P2 and S1P3 are widely expressed and regulate many diverse functions throughout the body. The S1P2 receptor, in particular, often exerts cellular functions which are opposed to the functions of the S1P1 receptor. As a consequence, antagonists at the S1P2 receptor have the potential to be useful in a contrasting context and different areas of indication compared to S1P1 antagonists. The present review will focus on the therapeutic potential of S1P2 receptor antagonists and discuss their opportunities as well as their potential risks. Open questions and areas which require further investigations will be emphasized in particular.
Collapse
Affiliation(s)
- Kira V Blankenbach
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Stephanie Schwalm
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Dagmar Meyer Zu Heringdorf
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| |
Collapse
|
27
|
Jiang L, Wang Y, Pan F, Zhao X, Zhang H, Lei M, Liu T, Lu JR. Synergistic effect of bioactive lipid and condition medium on cardiac differentiation of human mesenchymal stem cells from different tissues. Cell Biochem Funct 2016; 34:163-72. [PMID: 26990081 PMCID: PMC5031220 DOI: 10.1002/cbf.3175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/22/2016] [Accepted: 02/08/2016] [Indexed: 12/12/2022]
Abstract
Human umbilical cord mesenchymal stem cells (hUCMSCs) and human adipose tissue mesenchymal stem cells (hATMSCs) have the potential to differentiate into cardiomyocytes, making them promising therapeutic candidates for treating damaged cardiac tissues. Currently, however, the differentiated cells induced from hUCMSCs or hATMSCs can hardly display functional characteristics similar to cardiomyocytes. In this study, we have investigated the effects of bioactive lipid sphingosine-1-phosphate (S1P) on cardiac differentiations of hUCMSCs and hATMSCs in condition medium composed of cardiac myocytes culture medium or 5-azacytidine. Cardiac differentiations were identified through immunofluorescence staining, and the results were observed with fluorescence microscopy and confocal microscopy. Synergistic effects of S1P and condition medium on cell viability were evaluated by MTT assays. Functional characteristics similar to cardiomyocytes were evaluated through detecting calcium transient. The differentiated hUCMSCs or hATMSCs in each group into cardiomyocytes showed positive expressions of cardiac specific proteins, including α-actin, connexin-43 and myosin heavy chain-6 (MYH-6). MTT assays showed that suitable differentiation time was 14 days and that the optimal concentration of S1P was 0.5 μM. Moreover, incorporation of S1P and cardiac myocytes culture medium gave rise to calcium transients, an important marker for displaying in vivo electrophysiological properties. This feature was not observed in the S1P-5-azacytidine group, indicating the possible lack of cellular stimuli such as transforming growth factor-beta, TGF-β.
Collapse
Affiliation(s)
- Lili Jiang
- Dalian R&D Center for Stem Cell and Tissue Engineering, Faculty of Chemical Environmental and Biological Science and TechnologyDalian University of TechnologyDalianChina
| | - Yanwen Wang
- Cardiovascular and Genetic Medicine Research Groups, School of BiomedicineUniversity of ManchesterManchesterUK
| | - Fang Pan
- Biological Physics Group, School of Physics and AstronomyUniversity of ManchesterManchesterUK
| | - Xiubo Zhao
- Department of Chemical & Biological EngineeringUniversity of SheffieldMappin Street, Sheffield, S1 3JDUK
| | - Henggui Zhang
- Biological Physics Group, School of Physics and AstronomyUniversity of ManchesterManchesterUK
| | - Ming Lei
- Cardiovascular and Genetic Medicine Research Groups, School of BiomedicineUniversity of ManchesterManchesterUK
| | - Tianqing Liu
- Dalian R&D Center for Stem Cell and Tissue Engineering, Faculty of Chemical Environmental and Biological Science and TechnologyDalian University of TechnologyDalianChina
| | - Jian R. Lu
- Biological Physics Group, School of Physics and AstronomyUniversity of ManchesterManchesterUK
| |
Collapse
|
28
|
Tian T, Zhao Y, Huang Q, Li J. n-3 Polyunsaturated Fatty Acids Improve Inflammation via Inhibiting Sphingosine Kinase 1 in a Rat Model of Parenteral Nutrition and CLP-Induced Sepsis. Lipids 2016; 51:271-8. [PMID: 26856322 DOI: 10.1007/s11745-016-4129-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/21/2016] [Indexed: 12/31/2022]
Abstract
The sphingosine kinase 1 (SphK1)/sphingosine-1-phosphate (S1P) pathway plays a key role in inflammation. Parenteral nutrition containing n-3 polyunsaturated fatty acids (n-3 PUFA) may regulate inflammatory reactions. The aim of this study is to determine whether n-3 PUFA may improve inflammatory responses by neutralizing SphK1 signaling. Rat models of parenteral nutrition, cecal ligation and puncture (CLP)-induced sepsis were generated. Male Sprague-Dawley rats were operated for CLP on day 2 after venous catheterization. The rats were randomized to receive normal saline (NS; n = 20), parenteral nutrition (PN; n = 20), or PN + fish oil (FO; n = 20) for 5 days. The daily intake of fish oil (1.25-2.82 g EPA and 1.44-3.09 g DHA per 100 ml) in the FO group was approximately 1.8 g/kg body weight/day. Rats in the control group (n = 10) were subjected to sham operation and received a chow diet. Spleen tissues were collected for SphK1 and S1P receptor expression analysis. Our data showed that n-3 PUFA ameliorated the survival rate. SphK1 expression and its enzymatic activity were significantly upregulated in sepsis rats. Furthermore, mRNA and protein levels of S1PR3, but not S1PR1, were also facilitated after CLP. However, PN + FO dramatically decreased SphK1 mRNA level and its enzymatic activity. S1PR3 expression was also attenuated by FO addition. In conclusion, the anti-inflammatory effect of n-3 PUFA may be linked to the inhibition of the SphK1/S1P pathway in a rat model of parenteral nutrition and CLP-induced sepsis.
Collapse
Affiliation(s)
- Tao Tian
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Yunzhao Zhao
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| | - Qian Huang
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Jieshou Li
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| |
Collapse
|
29
|
Xing XQ, Li YL, Zhang YX, Xiao Y, Li ZD, Liu LQ, Zhou YS, Zhang HY, Liu YH, Zhang LH, Zhuang M, Chen YP, Ouyang SR, Wu XW, Yang J. Sphingosine kinase 1/sphingosine 1-phosphate signalling pathway as a potential therapeutic target of pulmonary hypertension. Int J Clin Exp Med 2015; 8:11930-5. [PMID: 26550106 DOI: pmid/26550106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/28/2015] [Indexed: 02/08/2023]
Abstract
Pulmonary hypertension is characterized by extensive vascular remodelling, leading to increased pulmonary vascular resistance and eventual death due to right heart failure. The pathogenesis of pulmonary hypertension involves vascular endothelial dysfunction and disordered vascular smooth muscle cell (VSMC) proliferation and migration, but the exact processes remain unknown. Sphingosine 1-phosphate (S1P) is a bioactive lysophospholipid involved in a wide spectrum of biological processes. S1P has been shown to regulate VSMC proliferation and migration and vascular tension via a family of five S1P G-protein-coupled receptors (S1P1-SIP5). S1P has been shown to have both a vasoconstrictive and vasodilating effect. The S1P receptors S1P1 and S1P3 promote, while S1P2 inhibits VSMC proliferation and migration in vitro in response to S1P. Moreover, it has been reported recently that sphingosine kinase 1 and S1P2 inhibitors might be useful therapeutic agents in the treatment of empirical pulmonary hypertension. The sphingosine kinase 1/S1P signalling pathways may play a role in the pathogenesis of pulmonary hypertension. Modulation of this pathway may offer novel therapeutic strategies.
Collapse
Affiliation(s)
- Xi-Qian Xing
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yan-Li Li
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yu-Xuan Zhang
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yi Xiao
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Zhi-Dong Li
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Li-Qiong Liu
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yu-Shan Zhou
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Hong-Yan Zhang
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yan-Hong Liu
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Li-Hui Zhang
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Min Zhuang
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yan-Ping Chen
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Sheng-Rong Ouyang
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Xu-Wei Wu
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Jiao Yang
- First Department of Respiratory Medicine, First Affiliated Hospital of Kunming Medical University Kunming, Yunnan, China
| |
Collapse
|
30
|
Althoff TF, Offermanns S. G-protein-mediated signaling in vascular smooth muscle cells — implications for vascular disease. J Mol Med (Berl) 2015; 93:973-81. [DOI: 10.1007/s00109-015-1305-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/14/2015] [Accepted: 06/02/2015] [Indexed: 10/24/2022]
|
31
|
Chen J, Tang H, Sysol JR, Moreno-Vinasco L, Shioura KM, Chen T, Gorshkova I, Wang L, Huang LS, Usatyuk PV, Sammani S, Zhou G, Raj JU, Garcia JGN, Berdyshev E, Yuan JXJ, Natarajan V, Machado RF. The sphingosine kinase 1/sphingosine-1-phosphate pathway in pulmonary arterial hypertension. Am J Respir Crit Care Med 2014; 190:1032-43. [PMID: 25180446 DOI: 10.1164/rccm.201401-0121oc] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Sphingosine kinases (SphKs) 1 and 2 regulate the synthesis of the bioactive sphingolipid sphingosine-1-phosphate (S1P), an important lipid mediator that promotes cell proliferation, migration, and angiogenesis. OBJECTIVES We aimed to examine whether SphKs and their product, S1P, play a role in the development of pulmonary arterial hypertension (PAH). METHODS SphK1(-/-), SphK2(-/-), and S1P lyase heterozygous (Sgpl1(+/-)) mice, a pharmacologic SphK inhibitor (SKI2), and a S1P receptor 2 (S1PR2) antagonist (JTE013) were used in rodent models of hypoxia-mediated pulmonary hypertension (HPH). S1P levels in lung tissues from patients with PAH and pulmonary arteries (PAs) from rodent models of HPH were measured. MEASUREMENTS AND MAIN RESULTS mRNA and protein levels of SphK1, but not SphK2, were significantly increased in the lungs and isolated PA smooth muscle cells (PASMCs) from patients with PAH, and in lungs of experimental rodent models of HPH. S1P levels were increased in lungs of patients with PAH and PAs from rodent models of HPH. Unlike SphK2(-/-) mice, SphK1(-/-) mice were protected against HPH, whereas Sgpl1(+/-) mice were more susceptible to HPH. Pharmacologic SphK1 and S1PR2 inhibition prevented the development of HPH in rodent models of HPH. Overexpression of SphK1 and stimulation with S1P potentially via ligation of S1PR2 promoted PASMC proliferation in vitro, whereas SphK1 deficiency inhibited PASMC proliferation. CONCLUSIONS The SphK1/S1P axis is a novel pathway in PAH that promotes PASMC proliferation, a major contributor to pulmonary vascular remodeling. Our results suggest that this pathway is a potential therapeutic target in PAH.
Collapse
Affiliation(s)
- Jiwang Chen
- 1 Section of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Smyth SS, Mueller P, Yang F, Brandon JA, Morris AJ. Arguing the case for the autotaxin-lysophosphatidic acid-lipid phosphate phosphatase 3-signaling nexus in the development and complications of atherosclerosis. Arterioscler Thromb Vasc Biol 2014; 34:479-86. [PMID: 24482375 DOI: 10.1161/atvbaha.113.302737] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The structurally simple glycero- and sphingo-phospholipids, lysophosphatidic acid (LPA) and sphingosine-1-phosphate, serve as important receptor-active mediators that influence blood and vascular cell function and are positioned to influence the events that contribute to the progression and complications of atherosclerosis. Growing evidence from preclinical animal models has implicated LPA, LPA receptors, and key enzymes involved in LPA metabolism in pathophysiologic events that may underlie atherosclerotic vascular disease. These observations are supported by genetic analysis in humans implicating a lipid phosphate phosphatase as a novel risk factor for coronary artery disease. In this review, we summarize current understanding of LPA production, metabolism, and signaling as may be relevant for atherosclerotic and other vascular disease.
Collapse
Affiliation(s)
- Susan S Smyth
- From the Veterans Affairs Medical Center, Cardiovascular Medicine Service, Lexington, KY (S.S.S., A.J.M.); and Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, KY (S.S.S., P.M., F.Y., J.A.B., A.J.M.)
| | | | | | | | | |
Collapse
|
33
|
Heilmann A, Schinke T, Bindl R, Wehner T, Rapp A, Haffner-Luntzer M, Liedert A, Amling M, Ignatius A. Systemic treatment with the sphingosine-1-phosphate analog FTY720 does not improve fracture healing in mice. J Orthop Res 2013; 31:1845-50. [PMID: 23818033 DOI: 10.1002/jor.22426] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 06/06/2013] [Indexed: 02/04/2023]
Abstract
Sphingosine-1-phosphate (S1P) has recently been recognized as a crucial coupling molecule of osteoclast and osteoblast activity provoking osteoanabolic effects. Targeting S1P receptors could, therefore, be a potential strategy to support bone formation in osteopenic diseases or in fracture repair. Here we investigated whether systemic treatment with the S1P analog FTY720 (Fingolimod) could improve fracture healing. Twelve-week-old, female C57BL/6 mice received an osteotomy of the femur, which was stabilized using an external fixator. The mice received a daily subcutaneous injection of either FTY720 (6 mg/kg) or vehicle from the third postoperative day. Fracture healing was evaluated after 10 and 21 days using biomechanical testing, µ-computed tomography, and histomorphometry. Because FTY720 is supposed to influence osteoclast recruitment, osteoclasts were identified in the fracture callus by staining for tartrate resistant acid phosphatase (TRAP). There were no significant differences in callus mechanical properties, tissue composition and osteoclast number between the groups, suggesting that systemically applied FTY720 did not influence bone regeneration in this model of regular fracture healing. Even if further studies should test the potency of FTY720 under unfavorable healing conditions, we conclude that the effect of systemically applied FTY720 on fracture healing might be inferior compared to other anabolic treatments. © 2013 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 31:1845-1850, 2013.
Collapse
Affiliation(s)
- Aline Heilmann
- Institute of Orthopaedic Research and Biomechanics, Center of Musculoskeletal Research, University of Ulm, Ulm, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kendig DM, Matsumoto AK, Moreland RS. Sphingosine-1-phosphate induced contraction of bladder smooth muscle. Eur J Pharmacol 2013; 720:355-62. [PMID: 24120660 DOI: 10.1016/j.ejphar.2013.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 10/02/2013] [Accepted: 10/03/2013] [Indexed: 11/29/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that contracts most smooth muscles. Although S1P has been shown to contract bladder smooth muscle, the mechanism(s) by which S1P initiates contraction has not been extensively investigated. The goal of this study was to determine if S1P-induced force generation and myosin light chain (MLC) phosphorylation are dependent on calcium sensitization pathways mediated by protein kinase C (PKC) and Rho kinase (ROCK) and which S1P receptor is important in this response. Bladder smooth muscle strips from rabbit and rat were mounted for isometric force recording and contracted in response to carbachol or S1P in the presence and absence of an inhibitor of PKC (3 µM Bisindolylmaleimide-1) or ROCK (1 µM H-1172). 10 µM S1P produced approximately 40% of the force generated in response to 110 mM KCl in rabbit bladder smooth muscle. S1P, up to 100 µM, did not produce a response in rat bladder smooth muscle, any response evoked was due to solvent (NaOH). S1P-dependent force development was associated with a concomitant increase in Ser(19), but not dual Thr(18)/Ser(19) MLC phosphorylation. Inhibition of PKC decreased force development, whereas inhibition of ROCK abolished S1P-induced force. An inhibitor of the S1P2 receptor, JTE-013, relaxed a S1P-induced contraction; whereas, an agonist with low affinity to the S1P2 receptor, dihydro-S1P, did not elicit a contraction. Our results suggest that S1P contracts rabbit, but not rat, bladder smooth muscle via the S1P2 receptor and is dependent on MLC phosphorylation and myofilament calcium sensitization primarily in response to ROCK activation.
Collapse
Affiliation(s)
- Derek M Kendig
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N 15th Street, MS #488, Philadelphia, PA 19102, USA
| | | | | |
Collapse
|
35
|
Tong S, Tian J, Wang H, Huang Z, Yu M, Sun L, Liu R, Liao M, Ning Z. H9N2 avian influenza infection altered expression pattern of sphiogosine-1-phosphate receptor 1 in BALB/c mice. Virol J 2013; 10:296. [PMID: 24073762 PMCID: PMC3849581 DOI: 10.1186/1743-422x-10-296] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 09/24/2013] [Indexed: 01/30/2023] Open
Abstract
Background The pathological damage inflicted by virulent AIV strains is often caused by inducing a positive feedback loop of cytokines in immune cells that cause excessive inflammation. Previous research has shown that a G protein-coupled receptor, sphingosine-1-phosphate receptor 1 (S1PR1), plays a crucial role in the development of excessive inflammation in influenza virus infection (Cell 146:861–862, 2011; Cell 146:980–991, 2011). BALB/c mice are common laboratory animals used in research of influenza virus; however the effects of influenza infections on expression patterns of S1PR1 in mice are unknown. Methods We investigated the expression patterns of S1PR1 in normal BALB/c mice and those infected by two distinct H9N2 AIV strains, one (A/chicken/Guangdong/V/2008,V) highly pathogenic, and the other (A/chicken/Guangdong/Ts/2004,Ts), non-pathogenic in mice, using quantitative PCR and immunohistochemistry (IHC) to detect S1PR1 mRNA and protein, respectively. Results S1PR1 mRNA was ubiquitously expressed in all the tissues examined, and significant differences were seen in mRNA expression between infected Ts, V and control mice in detected tissues, heart, liver, spleen, kidney and brain. S1PR1 protein was expressed in the cytoplasm and also demonstrated quantitative changes in expression in the various tissues between mice infected with the two strains of AIV. Conclusions Our results provided the first look at differences in S1PR1 expression patterns in BALB/c mice infected by non-pathogenic and highly pathogenic H9N2 influenza viruses. This information will not only be helpful in designing experiments to better understand the role of S1PR1 in virus-host interactions but also in developing novel anti-influenza agents to minimize the mortality and morbidity associated with highly virulent strains in avian and human populations.
Collapse
Affiliation(s)
- Shuang Tong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, People's Republic of China.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Vascular smooth muscle cells have attracted considerable interest as a model for a flexible program of gene expression. This cell type arises throughout the embryo body plan via poorly understood signaling cascades that direct the expression of transcription factors and microRNAs which, in turn, orchestrate the activation of contractile genes collectively defining this cell lineage. The discovery of myocardin and its close association with serum response factor has represented a major break-through for the molecular understanding of vascular smooth muscle cell differentiation. Retinoids have been shown to improve the outcome of vessel wall remodeling following injury and have provided further insights into the molecular circuitry that defines the vascular smooth muscle cell phenotype. This review summarizes the progress to date in each of these areas of vascular smooth muscle cell biology.
Collapse
|
37
|
Wang J, Yan CH, Li Y, Xu K, Tian XX, Peng CF, Tao J, Sun MY, Han YL. MicroRNA-31 controls phenotypic modulation of human vascular smooth muscle cells by regulating its target gene cellular repressor of E1A-stimulated genes. Exp Cell Res 2013; 319:1165-75. [PMID: 23518389 DOI: 10.1016/j.yexcr.2013.03.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 03/02/2013] [Accepted: 03/04/2013] [Indexed: 10/27/2022]
Abstract
Phenotypic modulation of vascular smooth muscle cells (VSMCs) plays a critical role in the pathogenesis of a variety of proliferative vascular diseases. The cellular repressor of E1A-stimulated genes (CREG) has been shown to play an important role in phenotypic modulation of VSMCs. However, the mechanism regulating CREG upstream signaling remains unclear. MicroRNAs (miRNAs) have recently been found to play a critical role in cell differentiation via target-gene regulation. This study aimed to identify a miRNA that binds directly to CREG, and may thus be involved in CREG-mediated VSMC phenotypic modulation. Computational analysis indicated that miR-31 bound to the CREG mRNA 3' untranslated region (3'-UTR). miR-31 was upregulated in quiescent differentiated VSMCs and downregulated in proliferative cells stimulated by platelet-derived growth factor and serum starvation, demonstrating a negative relationship with the VSMC differentiation marker genes, smooth muscle α-actin, calponin and CREG. Using gain-of-function and loss-of-function approaches, CREG and VSMC differentiation marker gene expression levels were shown to be suppressed by a miR-31 mimic, but increased by a miR-31 inhibitor at both protein and mRNA levels. Notably, miR-31 overexpression or inhibition affected luciferase expression driven by the CREG 3'-UTR containing the miR-31 binding site. Furthermore, miR-31-mediated VSMC phenotypic modulation was inhibited in CREG-knockdown human VSMCs. We also determined miR-31 levels in the serum of patients with coronary artery disease (CAD), with or without in stent restenosis and in healthy controls. miR-31 levels were higher in the serum of CAD patients with restenosis compared to CAD patients without restenosis and in healthy controls. In summary, these data demonstrate that miR-31 not only directly binds to its target gene CREG and modulates the VSMC phenotype through this interaction, but also can be an important biomarker in diseases involving VSMC phenotypic modulation. These novel findings may have extensive implications for the diagnosis and therapy of a variety of proliferative vascular diseases.
Collapse
Affiliation(s)
- Jie Wang
- Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Sphingosine-1-phosphate (S1P) regulates important functions in cardiac and vascular homeostasis. It has been implied to play causal roles in the pathogenesis of many cardiovascular disorders such as coronary artery disease, atherosclerosis, myocardial infarction, and heart failure. The majority of S1P in plasma is associated with high-density lipoproteins (HDL), and their S1P content has been shown to be responsible, at least in part, for several of the beneficial effects of HDL on cardiovascular risk. The attractiveness of S1P-based drugs for potential cardiovascular applications is increasing in the wake of the clinical approval of FTY720, but answers to important questions on the effects of S1P in cardiovascular biology and medicine must still be found. This chapter focuses on the current understanding of the role of S1P and its receptors in cardiovascular physiology, pathology, and disease.
Collapse
Affiliation(s)
- Bodo Levkau
- University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
39
|
Althoff TF, Albarrán Juárez J, Troidl K, Tang C, Wang S, Wirth A, Takefuji M, Wettschureck N, Offermanns S. Procontractile G protein-mediated signaling pathways antagonistically regulate smooth muscle differentiation in vascular remodeling. ACTA ACUST UNITED AC 2012; 209:2277-90. [PMID: 23129751 PMCID: PMC3501360 DOI: 10.1084/jem.20120350] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vascular smooth muscle (Sm) cells (VSMCs) are highly plastic. Their differentiation state can be regulated by serum response factor (SRF), which activates genes involved in Sm differentiation and proliferation by recruiting cofactors, such as members of the myocardin family and ternary complex factors (TCFs), respectively. However, the extracellular cues and upstream signaling mechanisms regulating SRF-dependent VSMC differentiation under in vivo conditions are poorly understood. In this study, we show that the procontractile signaling pathways mediated by the G proteins G(12)/G(13) and G(q)/G(11) antagonistically regulate VSMC plasticity in different models of vascular remodeling. In mice lacking Gα(12)/Gα(13) or their effector, the RhoGEF protein LARG, RhoA-dependent SRF-regulation was blocked and down-regulation of VSMC differentiation marker genes was enhanced. This was accompanied by an excessive vascular remodeling and exacerbation of atherosclerosis. In contrast, Sm-specific Gα(q)/Gα(11) deficiency blocked activation of extracellular signal-regulated kinase 1/2 and the TCF Elk-1, resulting in a reduced VSMC dedifferentiation in response to flow cessation or vascular injury. These data show that the balanced activity of both G protein-mediated pathways in VSMCs is required for an appropriate vessel remodeling response in vascular diseases and suggest new approaches to modulate Sm differentiation in vascular pathologies.
Collapse
Affiliation(s)
- Till F Althoff
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sefcik LS, Petrie Aronin CE, Botchwey EA. Engineering vascularized tissues using natural and synthetic small molecules. Organogenesis 2012; 4:215-27. [PMID: 19337401 DOI: 10.4161/org.4.4.6963] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Accepted: 09/10/2008] [Indexed: 12/21/2022] Open
Abstract
Vascular growth and remodeling are complex processes that depend on the proper spatial and temporal regulation of many different signaling molecules to form functional vascular networks. The ability to understand and regulate these signals is an important clinical need with the potential to treat a wide variety of disease pathologies. Current approaches have focused largely on the delivery of proteins to promote neovascularization of ischemic tissues, most notably VEGF and FGF. Although great progress has been made in this area, results from clinical trials are disappointing and safer and more effective approaches are required. To this end, biological agents used for therapeutic neovascularization must be explored beyond the current well-investigated classes. This review focuses on potential pathways for novel drug discovery, utilizing small molecule approaches to induce and enhance neovascularization. Specifically, four classes of new and existing molecules are discussed, including transcriptional activators, receptor selective agonists and antagonists, natural product-derived small molecules, and novel synthetic small molecules.
Collapse
Affiliation(s)
- Lauren S Sefcik
- Department of Biomedical Engineering; and Department of Orthopaedic Surgery; University of Virginia; Charlottesville, Virginia USA; Center for Immunity, Inflammation and Regenerative Medicine (CIIR); University of Virginia; Charlottesville, Virginia USA
| | | | | |
Collapse
|
41
|
The Reduction of Allograft Arteriosclerosis in Intestinal Transplant Is Associated With Sphingosine Kinase 1/Sphingosine-1-Phosphate Signaling After Fish Oil Treatment. Transplantation 2012; 93:989-96. [DOI: 10.1097/tp.0b013e31824d709d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
42
|
Meissner A, Yang J, Kroetsch JT, Sauvé M, Dax H, Momen A, Noyan-Ashraf MH, Heximer S, Husain M, Lidington D, Bolz SS. Tumor necrosis factor-α-mediated downregulation of the cystic fibrosis transmembrane conductance regulator drives pathological sphingosine-1-phosphate signaling in a mouse model of heart failure. Circulation 2012; 125:2739-50. [PMID: 22534621 DOI: 10.1161/circulationaha.111.047316] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Sphingosine-1-phosphate (S1P) signaling is a central regulator of resistance artery tone. Therefore, S1P levels need to be tightly controlled through the delicate interplay of its generating enzyme sphingosine kinase 1 and its functional antagonist S1P phosphohydrolase-1. The intracellular localization of S1P phosphohydrolase-1 necessitates the import of extracellular S1P into the intracellular compartment before its degradation. The present investigation proposes that the cystic fibrosis transmembrane conductance regulator transports extracellular S1P and hence modulates microvascular S1P signaling in health and disease. METHODS AND RESULTS In cultured murine vascular smooth muscle cells in vitro and isolated murine mesenteric and posterior cerebral resistance arteries ex vivo, the cystic fibrosis transmembrane conductance regulator (1) is critical for S1P uptake; (2) modulates S1P-dependent responses; and (3) is downregulated in vitro and in vivo by tumor necrosis factor-α, with significant functional consequences for S1P signaling and vascular tone. In heart failure, tumor necrosis factor-α downregulates the cystic fibrosis transmembrane conductance regulator across several organs, including the heart, lung, and brain, suggesting that it is a fundamental mechanism with implications for systemic S1P effects. CONCLUSIONS We identify the cystic fibrosis transmembrane conductance regulator as a critical regulatory site for S1P signaling; its tumor necrosis factor-α-dependent downregulation in heart failure underlies an enhancement in microvascular tone. This molecular mechanism potentially represents a novel and highly strategic therapeutic target for cardiovascular conditions involving inflammation.
Collapse
Affiliation(s)
- Anja Meissner
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Differential expression of sphingosine-1-phosphate receptors in abdominal aortic aneurysms. Mediators Inflamm 2012; 2012:643609. [PMID: 22547907 PMCID: PMC3323867 DOI: 10.1155/2012/643609] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/19/2012] [Indexed: 11/17/2022] Open
Abstract
Objective. Inflammation plays a key role in the pathophysiology of abdominal aortic aneurysms (AAAs). Newly discovered Sphingosine-1-Phosphate Receptors (S1P receptors) are critical in modulating inflammatory response via prostaglandin production. The aim of the current study was to investigate the expression of different S1P receptors in AAAs and compared with normal aortas at the protein level. Materials and Methods. Aortic specimens were harvested during aortic reconstructive surgery for the AAA group or during organ transplant for the control group. The protein expression of S1P1, 2 and 3 in AAAs and normal aortas was assessed by Western blotting and immunohistochemical analysis. Results. There were 40 AAAs and 20 control aortas collected for the receptor analysis. For Western blot analysis, S1P1 expression was not detected in either group; S1P2 protein was constitutively detected in both types of aortas but its expression level was significantly decreased
by 73% (P < 0.05) in AAAs compared with the control group. In contrast, strong S1P3 expression was detected in AAAs aortas but not in normal aortas. Immumohistochemical staining showed
similar results, except a weak S1P3 signal was detectable in normal aortas. Conclusions. Western blot and staining results consistently showed the down-regulation of the S1P2 protein with simultaneous up-regulation of the S1P3 protein in AAAs. Since those newly discovered receptors play an important role in the inflammatory cascade, the modulating of S1P signaling, particularly via S1P2 and S1P3, could represent novel therapeutic targets in future AAA treatments.
Collapse
|
44
|
Duru EA, Fu Y, Davies MG. Role of S-1-P receptors and human vascular smooth muscle cell migration in diabetes and metabolic syndrome. J Surg Res 2012; 177:e75-82. [PMID: 22480845 DOI: 10.1016/j.jss.2011.12.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Revised: 11/18/2011] [Accepted: 12/06/2011] [Indexed: 11/25/2022]
Abstract
BACKGROUND Sphingosine-1-phosphate (S-1-P) is a bioactive sphingolipid released from activated platelets that stimulates migration of vascular smooth muscle cells (VSMC) in vitro. S-1-P is associated with oxidized low-density lipoprotein (oxLDL) and is important in vessel remodeling. S-1-P will activate multiple G protein-coupled receptors (S-1-PR 1 to 5), which can regulate multiple cellular functions, including cell migration. The aim of this study is to examine the role of S-1-PR signaling during smooth muscle cell migration in response to S-1-P. METHODS Human VSMCs were cultured in vitro. Expression of S-1-PR 1 to 5 was determined in conditions mirroring diabetes (40 mM glucose) and metabolic syndrome (25 mM glucose with 20 μM linoleic acid and 20 μM oleic acid). Linear wound and Boyden microchemotaxis assays of migration were performed in the presence of S-1-P with and without siRNA against S-1-PR 1 to 5. Assays were performed for activation of ERK1/2, p38(MAPK) and JNK. RESULTS Human VSMCs express S-1-PR1, S-1-PR2, and S-1-PR3. There was no significant expression of S-1-PR4 and S-1-PR5. The expression of S-1-PR1 and S-1-PR3 is enhanced under high glucose conditions and metabolic syndrome conditions. Migration of VSMC in response to S-1-P is enhanced 2-fold by diabetes and 4-fold by metabolic syndrome. In diabetes, S-1-PR1 expression is enhanced, while S-1-PR2 and S-1-PR3 expression are both maintained. In metabolic syndrome, S-1-PR1 and 3 expressions are enhanced and that of S-1-PR2 is reduced. siRNA to S-1-PR1 results in a 2-fold reduction in S-1-P-mediated cell migration under all conditions. siRNA to S-1-PR2 enhanced cell migration only under normal conditions, while siRNA S-1-PR3 decreased migration in metabolic syndrome only. Down-regulation of S-1-PR1 reduced ERK1/2 activation in response to S-1-P, while that of S-1-PR2 had no effect under normal conditions. In diabetes, down-regulation of S-1-PR1 reduced activation of all three MAPKs. In metabolic syndrome, down-regulation of S-1-PR1 and S-1-PR3 reduced activation of all three MAPKs. CONCLUSION S-1-PR 1, 2, and 3 regulate human VSMC migration and their expression level and function are modulated by conditions simulating diabetes and metabolic syndrome.
Collapse
Affiliation(s)
- Enrico A Duru
- Vascular Biology and Therapeutics Program, The Methodist Hospital Research Institute, Houston, TX, USA
| | | | | |
Collapse
|
45
|
Shimizu T, De Wispelaere A, Winkler M, D'Souza T, Caylor J, Chen L, Dastvan F, Deou J, Cho A, Larena-Avellaneda A, Reidy M, Daum G. Sphingosine-1-phosphate receptor 3 promotes neointimal hyperplasia in mouse iliac-femoral arteries. Arterioscler Thromb Vasc Biol 2012; 32:955-61. [PMID: 22308044 DOI: 10.1161/atvbaha.111.241034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The objective of this study was to define a role for sphingosine-1-phosphate receptor 3 (S1PR3) in intimal hyperplasia. METHODS AND RESULTS A denudation model of the iliac-femoral artery in wild-type and S1PR3-null mice was used to define a role for S1PR3 in the arterial injury response because we found in humans and mice that expression of S1PR3 was higher in these arteries compared with carotid arteries. At 28 days after surgery, wild-type arteries formed significantly larger lesions than S1PR3-null arteries. Bromodeoxyuridine labeling experiments demonstrated that on injury, wild-type arteries exhibited higher medial as well as intimal proliferation than S1PR3-null arteries. Because S1PR3 expression in vitro was low, we expressed S1PR3 in S1PR3-null smooth muscle cells (SMCs) using retroviral-mediated gene transfer to study the effects of S1PR3 on cell functions and signaling. SMCs expressing S1PR3, but not vector-transfected controls, responded to sphingosine-1-phosphate stimulation with activation of Rac, Erk, and Akt. SMCs expressing S1PR3 also migrated more. CONCLUSIONS In humans and mice, S1PR3 expression was higher in iliac-femoral arteries compared with carotid arteries. S1PR3 promoted neointimal hyperplasia on denudation of iliac-femoral arteries in mice, likely by stimulating cell migration and proliferation through activation of signaling pathways involving Erk, Akt, and Rac.
Collapse
Affiliation(s)
- Takuya Shimizu
- Department of Pathology, University of Washington, Seattle, 98109, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lee MY, Garvey SM, Ripley ML, Wamhoff BR. Genome-wide microarray analyses identify the protein C receptor as a novel calcineurin/nuclear factor of activated T cells-dependent gene in vascular smooth muscle cell phenotypic modulation. Arterioscler Thromb Vasc Biol 2012; 31:2665-75. [PMID: 21903947 DOI: 10.1161/atvbaha.111.235960] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Calcineurin (Cn) and the nuclear factor of activated T cells (NFAT) family of transcription factors are critical in vascular smooth muscle cell (SMC) development and pathology. Here, we used a genomics approach to identify and validate NFAT gene targets activated during platelet-derived growth factor-BB (PDGF-BB)-induced SMC phenotypic modulation. METHODS AND RESULTS Genome-wide expression arrays were used to identify genes both (1) differentially activated in response to PDGF-BB and (2) whose differential expression was reduced by both the Cn inhibitor cyclosporin A and the NFAT inhibitor A-285222. The 20 most pharmacologically sensitive genes were validated by quantitative reverse transcription-polymerase chain reaction analysis of PDGF-BB-stimulated SMCs in the presence of Cn/NFAT inhibitors, including the VIVIT peptide. In all experiments, protein C receptor (PROCR) gene activation was reduced. We showed that PROCR expression was virtually absent in untreated, quiescent SMCs. PDGF-BB stimulation, however, induced significant PROCR promoter activation and downstream protein expression in a Cn/NFAT-dependent manner. Mutation of a species-conserved, NFAT binding motif significantly attenuated PDGF-BB-induced PROCR promoter activity, thereby distinguishing NFAT as the first PROCR transcriptional activator to date. Moreover, SMC PROCR expression was upregulated in the neointima as early as 7 days following acute vascular injury in rat carotid arteries. CONCLUSION We hereby report PROCR as a novel, NFAT-dependent gene that may be implicated in vascular restenosis and consequent inward remodeling.
Collapse
Affiliation(s)
- Monica Y Lee
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
47
|
Du J, Zeng C, Li Q, Chen B, Liu H, Huang X, Huang Q. LPS and TNF-α induce expression of sphingosine-1-phosphate receptor-2 in human microvascular endothelial cells. Pathol Res Pract 2012; 208:82-8. [PMID: 22244964 DOI: 10.1016/j.prp.2011.11.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 10/18/2011] [Accepted: 11/13/2011] [Indexed: 01/04/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sophospholipid with various S1P receptor (S1PR) expression profiles in cells of different origin. S1PR1, R3 and - to a lesser extent - R2 were the main receptors expressed in most of endothelial cells (ECs). The balances in the expression and activation of S1PR1, R2 and R3 help to maintain the physiological functions of ECs. Reverse transcription-PCR and Western blotting were used to detect the mRNA transcript level and protein expression of S1PR. Endothelial barrier function was measured by transflux of tracer protein through endothelial monolayer. Human dermal microvascular ECs predominantly expressed S1PR1 and S1PR3. Lipopolysaccharide (LPS) or tumor necrosis factor-α (TNF-α) significantly upregulated S1PR2 mRNA and protein levels. The application of S1PR2 antagonist JTE-013 decreased the endothelial monolayer hyper-permeability response induced by LPS and TNF-α. Inflammatory mediators LPS and TNF-α induce S1PR2 expression in endothelium, suggesting that S1PR2 up-regulation may be involved in LPS and TNF-α elicited endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Jing Du
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou 510515, PR China
| | | | | | | | | | | | | |
Collapse
|
48
|
Mousseau Y, Mollard S, Faucher-Durand K, Richard L, Nizou A, Cook-Moreau J, Baaj Y, Qiu H, Plainard X, Fourcade L, Funalot B, Sturtz FG. Fingolimod potentiates the effects of sunitinib malate in a rat breast cancer model. Breast Cancer Res Treat 2011; 134:31-40. [PMID: 22160641 DOI: 10.1007/s10549-011-1903-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 11/24/2011] [Indexed: 02/04/2023]
Abstract
Most of the antiangiogenic strategies used in oncology principally target endothelial cells through the vascular endothelial growth factor (VEGF) pathway. Multiple kinase inhibitors can secondarily reduce mural cell stabilization of the vessels by blocking platelet-derived growth factor receptor (PDGFR) activity. However, sphingosine-1-phosphate (S1P), which is also implicated in mural cell recruitment, has yet to be targeted in clinical practice. We therefore investigated the potential of a simultaneous blockade of the PDGF and S1P pathways on the chemotactic responses of vascular smooth muscle cells (VSMCs) and the resulting effects of this blockade on breast tumor growth. Due to crosstalk between the S1P and PDGF pathways, we used AG1296 and/or VPC-23019 to inhibit PDGFR-β and S1PR1/S1PR3 receptors, respectively. We showed that S1PR1 and S1PR3 are the principal receptors that mediate the S1P chemotactic signal on rat VSMCs and that they act synergistically with PDGFR-β during PDGF-B signaling. We also showed that simultaneous blockade of the PDGFR-β and S1PR1/S1PR3 signals had a synergistic effect, decreasing VSMC migration velocity toward endothelial cell and breast carcinoma cell-secreted cytokines by 65-90%. This blockade also strongly decreased the ability of VSMCs to form a three-dimensional cell network. Similar results were obtained with the combination of sunitinib malate (a VEGFR/PDGFR kinase inhibitor) and fingolimod (an S1P analog). Sunitinib malate is a clinically approved cancer treatment, whereas fingolimod is currently indicated only for treatment of multiple sclerosis. Orally administered, the combination of these drugs greatly decreased rat breast tumor growth in a syngeneic cancer model (Walker 256). This bi-therapy did not exert cumulative toxicity and histological analysis of the tumors revealed normalization of the tumor vasculature. The simultaneous blockade of these signaling pathways with sunitinib malate and fingolimod may provide an effective means of reducing tumor angiogenesis, and may improve the delivery of other chemotherapies.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Aorta, Thoracic/pathology
- Carcinoma 256, Walker/blood supply
- Carcinoma 256, Walker/drug therapy
- Carcinoma 256, Walker/pathology
- Cell Movement
- Cells, Cultured
- Drug Screening Assays, Antitumor
- Drug Synergism
- Female
- Fingolimod Hydrochloride
- Indoles/administration & dosage
- Male
- Mammary Neoplasms, Experimental/blood supply
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Neoplasm Transplantation
- Propylene Glycols/administration & dosage
- Proto-Oncogene Proteins c-sis/pharmacology
- Proto-Oncogene Proteins c-sis/physiology
- Pyrroles/administration & dosage
- Rats
- Rats, Sprague-Dawley
- Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptors, Lysosphingolipid/metabolism
- Receptors, Lysosphingolipid/physiology
- Sphingosine/administration & dosage
- Sphingosine/analogs & derivatives
- Sphingosine-1-Phosphate Receptors
- Statistics, Nonparametric
- Sunitinib
- Tumor Burden/drug effects
- Tyrphostins/pharmacology
Collapse
Affiliation(s)
- Yoanne Mousseau
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Limoges, Limoges, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Schuchardt M, Tölle M, Prüfer J, van der Giet M. Pharmacological relevance and potential of sphingosine 1-phosphate in the vascular system. Br J Pharmacol 2011; 163:1140-62. [PMID: 21309759 DOI: 10.1111/j.1476-5381.2011.01260.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) was identified as a crucial molecule for regulating immune responses, inflammatory processes as well as influencing the cardiovascular system. S1P mediates differentiation, proliferation and migration during vascular development and homoeostasis. S1P is a naturally occurring lipid metabolite and is present in human blood in nanomolar concentrations. S1P is not only involved in physiological but also in pathophysiological processes. Therefore, this complex signalling system is potentially interesting for pharmacological intervention. Modulation of the system might influence inflammatory, angiogenic or vasoregulatory processes. S1P activates G-protein coupled receptors, namely S1P(1-5) , whereas only S1P(1-3) is present in vascular cells. S1P can also act as an intracellular signalling molecule. This review highlights the pharmacological potential of S1P signalling in the vascular system by giving an overview of S1P-mediated processes in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). After a short summary of S1P metabolism and signalling pathways, the role of S1P in EC and VSMC proliferation and migration, the cause of relaxation and constriction of arterial blood vessels, the protective functions on endothelial apoptosis, as well as the regulatory function in leukocyte adhesion and inflammatory responses are summarized. This is followed by a detailed description of currently known pharmacological agonists and antagonists as new tools for mediating S1P signalling in the vasculature. The variety of effects influenced by S1P provides plenty of therapeutic targets currently under investigation for potential pharmacological intervention.
Collapse
Affiliation(s)
- Mirjam Schuchardt
- Charité- Universitätsmedizin Berlin, CharitéCentrum 10, Department of Nephrology, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin, Germany
| | | | | | | |
Collapse
|
50
|
Mack CP. Signaling mechanisms that regulate smooth muscle cell differentiation. Arterioscler Thromb Vasc Biol 2011; 31:1495-505. [PMID: 21677292 DOI: 10.1161/atvbaha.110.221135] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Extensive studies over the last 30 years have demonstrated that vascular smooth muscle cell (SMC) differentiation and phenotypic modulation is controlled by a dynamic array of environmental cues. The identification of the signaling mechanisms by which these environmental cues regulate SMC phenotype has been more difficult because of our incomplete knowledge of the transcription mechanisms that regulate SMC-specific gene expression. However, recent advances in this area have provided significant insight, and the goal of this review is to summarize the signaling mechanisms by which extrinsic cues control SMC differentiation.
Collapse
Affiliation(s)
- Christopher P Mack
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599-7525, USA.
| |
Collapse
|