1
|
Matsuno A, Sumida H, Nakanishi H, Ikeyama Y, Ishii T, Omori I, Saito H, Iwasawa O, Sugimori A, Yoshizaki A, Katoh H, Ishikawa S, Sato S. Keratinocyte proline-rich protein modulates immune and epidermal response in imiquimod-induced psoriatic skin inflammation. Exp Dermatol 2023; 32:2121-2130. [PMID: 37926955 DOI: 10.1111/exd.14960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
Psoriasis is a persistent inflammatory skin disease thought to arise as a result of the infiltration of inflammatory cells and activation of keratinocytes. Recent advances in basic research and clinical experience revealed that the interleukin (IL)-23/IL-17 axis has been identified as a major immune pathway in psoriasis. However, it remains unclear how keratinocyte factors contribute to the pathology of psoriasis. Keratinocyte proline-rich protein (KPRP) is a proline-rich insoluble protein, which is present in the epidermis and is likely to be involved in the skin barrier function. Here, to investigate the potential roles of KPRP in psoriatic skin inflammation, Kprp-modified mice were applied in the imiquimod (IMQ)-induced skin inflammation model, which develops psoriasis-like epidermal hyperplasia and cutaneous inflammation features. Then, heterozygous knockout (Kprp+/- ) but not homozygous knockout (Kprp-/- ) mice displayed attenuated skin erythema compared to control wild-type mice. In addition, RNA sequencing, quantitative PCR and/or histological analysis detected changes in the expression of several molecules related to psoriatic inflammation or keratinocyte differentiation in Kprp+/- mice, but not Kprp-/- mice. Further analysis exhibited reduced IL-17-producing γδlow T cells and amplified epidermal hyperplasia in Kprp+/- mice, which were implied to be related to decreased expression of β-defensins and increased expression of LPAR1 (Lysophosphatidic acid receptor 1), respectively. Thus, our results imply that KPRP has the potential as a therapeutic target in psoriatic skin inflammation.
Collapse
Affiliation(s)
- Ai Matsuno
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hayakazu Sumida
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Scleroderma Center, The University of Tokyo Hospital, Tokyo, Japan
- SLE Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Hirofumi Nakanishi
- Research and Development Division, Rohto Pharmaceutical Company, Osaka, Japan
| | - Yoshifumi Ikeyama
- Research and Development Division, Rohto Pharmaceutical Company, Osaka, Japan
| | - Tsuyoshi Ishii
- Research and Development Division, Rohto Pharmaceutical Company, Osaka, Japan
| | - Issei Omori
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hinako Saito
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Okuto Iwasawa
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ayaka Sugimori
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
2
|
Dacheux MA, Norman DD, Tigyi GJ, Lee SC. Emerging roles of lysophosphatidic acid receptor subtype 5 (LPAR5) in inflammatory diseases and cancer. Pharmacol Ther 2023; 245:108414. [PMID: 37061203 DOI: 10.1016/j.pharmthera.2023.108414] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid mediator that regulates a variety of cellular functions such as cell proliferation, migration, survival, calcium mobilization, cytoskeletal rearrangements, and neurite retraction. The biological actions of LPA are mediated by at least six G protein-coupled receptors known as LPAR1-6. Given that LPAR1-3 were among the first LPARs identified, the majority of research efforts have focused on understanding their biology. This review provides an in-depth discussion of LPAR5, which has recently emerged as a key player in regulating normal intestinal homeostasis and modulating pathological conditions such as pain, itch, inflammatory diseases, and cancer. We also present a chronological overview of the efforts made to develop compounds that target LPAR5 for use as tool compounds to probe or validate LPAR5 biology and therapeutic agents for the treatment of inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Mélanie A Dacheux
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States of America
| | - Derek D Norman
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States of America
| | - Gábor J Tigyi
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States of America
| | - Sue Chin Lee
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States of America.
| |
Collapse
|
3
|
Pei J, Cai L, Wang F, Xu C, Pei S, Guo H, Sun X, Chun J, Cong X, Zhu W, Zheng Z, Chen X. LPA 2 Contributes to Vascular Endothelium Homeostasis and Cardiac Remodeling After Myocardial Infarction. Circ Res 2022; 131:388-403. [PMID: 35920162 DOI: 10.1161/circresaha.122.321036] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Myocardial infarction (MI) is one of the most dangerous adverse cardiovascular events. Our previous study found that lysophosphatidic acid (LPA) is increased in human peripheral blood after MI, and LPA has a protective effect on the survival and proliferation of various cell types. However, the role of LPA and its receptors in MI is less understood. OBJECTIVES To study the unknown role of LPA and its receptors in heart during MI. METHODS AND RESULTS In this study, we found that mice also had elevated LPA level in peripheral blood, as well as increased cardiac expression of its receptor LPA2 in the early stages after MI. With adult and neonate MI models in global Lpar2 knockout (Lpar2-KO) mice, we found Lpar2 deficiency increased vascular leak leading to disruption of its homeostasis, so as to impaired heart function and increased early mortality. Histological examination revealed larger scar size, increased fibrosis, and reduced vascular density in the heart of Lpar2-KO mice. Furthermore, Lpar2-KO also attenuated blood flow recovery after femoral artery ligation with decreased vascular density in gastrocnemius. Our study revealed that Lpar2 was mainly expressed and altered in cardiac endothelial cells during MI, and use of endothelial-specific Lpar2 knockout mice phenocopied the global knockout mice. Additionally, adenovirus-Lpar2 and pharmacologically activated LPA2 significantly improved heart function, reduced scar size, increased vascular formation, and alleviated early mortality by maintaining vascular homeostasis owing to protecting vessels from leakage. Mechanistic studies demonstrated that LPA-LPA2 signaling could promote endothelial cell proliferation through PI3K-Akt/PLC-Raf1-Erk pathway and enhanced endothelial cell tube formation via PKD1-CD36 signaling. CONCLUSIONS Our results indicate that endothelial LPA-LPA2 signaling promotes angiogenesis and maintains vascular homeostasis, which is vital for restoring blood flow and repairing tissue function in ischemic injuries. Targeting LPA-LPA2 signal might have clinical therapeutic potential to protect the heart from ischemic injury.
Collapse
Affiliation(s)
- Jianqiu Pei
- State Key Laboratory of Cardiovascular Disease (J.P., L.C., C.X., S.P., X.C., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, China (J.P., Z.Z.)
| | - Lin Cai
- State Key Laboratory of Cardiovascular Disease (J.P., L.C., C.X., S.P., X.C., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China (L.C.)
| | - Fang Wang
- State Key Laboratory of Cardiovascular Disease, Center of Laboratory Medicine (F.W., X. Cong, X. Chen), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chuansheng Xu
- State Key Laboratory of Cardiovascular Disease (J.P., L.C., C.X., S.P., X.C., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shengqiang Pei
- State Key Laboratory of Cardiovascular Disease (J.P., L.C., C.X., S.P., X.C., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongwei Guo
- Department of Cardiovascular Surgery (H.G., X.S., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaogang Sun
- Department of Cardiovascular Surgery (H.G., X.S., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA (J.C.)
| | - Xiangfeng Cong
- State Key Laboratory of Cardiovascular Disease, Center of Laboratory Medicine (F.W., X. Cong, X. Chen), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiquan Zhu
- Department of Medicine, Program in Molecular Medicine, Department of Internal Medicine, Division of Cardiovascular Medicine, Department of Pathology, University of Utah, Salt Lake City (W.Z.)
| | - Zhe Zheng
- Department of Cardiovascular Surgery (H.G., X.S., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, China (J.P., Z.Z.)
| | - Xi Chen
- State Key Laboratory of Cardiovascular Disease (J.P., L.C., C.X., S.P., X.C., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Cardiovascular Disease, Center of Laboratory Medicine (F.W., X. Cong, X. Chen), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Qiu Y, Shen J, Jiang W, Yang Y, Liu X, Zeng Y. Sphingosine 1-phosphate and its regulatory role in vascular endothelial cells. Histol Histopathol 2022; 37:213-225. [PMID: 35118637 DOI: 10.14670/hh-18-428] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive metabolite of sphingomyelin. S1P activates a series of signaling cascades by acting on its receptors S1PR1-3 on endothelial cells (ECs), which plays an important role in endothelial barrier maintenance, anti-inflammation, antioxidant and angiogenesis, and thus is considered as a potential therapeutic biomarker for ischemic stroke, sepsis, idiopathic pulmonary fibrosis, cancers, type 2 diabetes and cardiovascular diseases. We presently review the levels of S1P in those vascular and vascular-related diseases. Plasma S1P levels were reduced in various inflammation-related diseases such as atherosclerosis and sepsis, but were increased in other diseases including type 2 diabetes, neurodegeneration, cerebrovascular damages such as acute ischemic stroke, Alzheimer's disease, vascular dementia, angina, heart failure, idiopathic pulmonary fibrosis, community-acquired pneumonia, and hepatocellular carcinoma. Then, we highlighted the molecular mechanism by which S1P regulated EC biology including vascular development and angiogenesis, inflammation, permeability, and production of reactive oxygen species (ROS), nitric oxide (NO) and hydrogen sulfide (H₂S), which might provide new ways for exploring the pathogenesis and implementing individualized therapy strategies for those diseases.
Collapse
Affiliation(s)
- Yan Qiu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Junyi Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yi Yang
- Department of Orthopeadics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Duflot T, Tu L, Leuillier M, Messaoudi H, Groussard D, Feugray G, Azhar S, Thuillet R, Bauer F, Humbert M, Richard V, Guignabert C, Bellien J. Preventing the Increase in Lysophosphatidic Acids: A New Therapeutic Target in Pulmonary Hypertension? Metabolites 2021; 11:metabo11110784. [PMID: 34822442 PMCID: PMC8621392 DOI: 10.3390/metabo11110784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/11/2021] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of premature death and disability in humans that are closely related to lipid metabolism and signaling. This study aimed to assess whether circulating lysophospholipids (LPL), lysophosphatidic acids (LPA) and monoacylglycerols (MAG) may be considered as potential therapeutic targets in CVD. For this objective, plasma levels of 22 compounds (13 LPL, 6 LPA and 3 MAG) were monitored by liquid chromatography coupled with tandem mass spectrometry (HPLC/MS2) in different rat models of CVD, i.e., angiotensin-II-induced hypertension (HTN), ischemic chronic heart failure (CHF) and sugen/hypoxia(SuHx)-induced pulmonary hypertension (PH). On one hand, there were modest changes on the monitored compounds in HTN (LPA 16:0, 18:1 and 20:4, LPC 16:1) and CHF (LPA 16:0, LPC 18:1 and LPE 16:0 and 18:0) models compared to control rats but these changes were no longer significant after multiple testing corrections. On the other hand, PH was associated with important changes in plasma LPA with a significant increase in LPA 16:0, 18:1, 18:2, 20:4 and 22:6 species. A deleterious impact of LPA was confirmed on cultured human pulmonary smooth muscle cells (PA-SMCs) with an increase in their proliferation. Finally, plasma level of LPA(16:0) was positively associated with the increase in pulmonary artery systolic pressure in patients with cardiac dysfunction. This study demonstrates that circulating LPA may contribute to the pathophysiology of PH. Additional experiments are needed to assess whether the modulation of LPA signaling in PH may be of interest.
Collapse
Affiliation(s)
- Thomas Duflot
- UNIROUEN, INSERM U1096, CHU Rouen, Department of Pharmacology, Normandie University, F-76000 Rouen, France; (V.R.); (J.B.)
- Correspondence: ; Tel.: +33-2-32-88-84-91
| | - Ly Tu
- INSERM UMR_S 999, Hôpital Marie Lannelongue, F-92350 Le Plessis-Robinson, France; (L.T.); (R.T.); (M.H.); (C.G.)
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, F-92290 Châtenay-Malabry, France
| | - Matthieu Leuillier
- UNIROUEN, INSERM U1096, Normandie University, F-76000 Rouen, France; (M.L.); (H.M.); (D.G.); (S.A.)
| | - Hind Messaoudi
- UNIROUEN, INSERM U1096, Normandie University, F-76000 Rouen, France; (M.L.); (H.M.); (D.G.); (S.A.)
| | - Déborah Groussard
- UNIROUEN, INSERM U1096, Normandie University, F-76000 Rouen, France; (M.L.); (H.M.); (D.G.); (S.A.)
| | - Guillaume Feugray
- UNIROUEN, INSERM U1096, CHU Rouen, Department of General Biochemistry, Normandie University, F-76000 Rouen, France;
| | - Saïda Azhar
- UNIROUEN, INSERM U1096, Normandie University, F-76000 Rouen, France; (M.L.); (H.M.); (D.G.); (S.A.)
| | - Raphaël Thuillet
- INSERM UMR_S 999, Hôpital Marie Lannelongue, F-92350 Le Plessis-Robinson, France; (L.T.); (R.T.); (M.H.); (C.G.)
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, F-92290 Châtenay-Malabry, France
| | - Fabrice Bauer
- UNIROUEN, INSERM U1096, CHU Rouen, Department of Cardiology, Normandie University, F-76000 Rouen, France;
| | - Marc Humbert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, F-92350 Le Plessis-Robinson, France; (L.T.); (R.T.); (M.H.); (C.G.)
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, F-92290 Châtenay-Malabry, France
| | - Vincent Richard
- UNIROUEN, INSERM U1096, CHU Rouen, Department of Pharmacology, Normandie University, F-76000 Rouen, France; (V.R.); (J.B.)
| | - Christophe Guignabert
- INSERM UMR_S 999, Hôpital Marie Lannelongue, F-92350 Le Plessis-Robinson, France; (L.T.); (R.T.); (M.H.); (C.G.)
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, F-92290 Châtenay-Malabry, France
| | - Jérémy Bellien
- UNIROUEN, INSERM U1096, CHU Rouen, Department of Pharmacology, Normandie University, F-76000 Rouen, France; (V.R.); (J.B.)
| |
Collapse
|
6
|
Antimicrobial and Immunomodulatory Activity of Herb Extracts Used in Burn Wound Healing: "San Huang Powder". EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2900060. [PMID: 34675981 PMCID: PMC8526243 DOI: 10.1155/2021/2900060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/07/2021] [Accepted: 09/11/2021] [Indexed: 12/20/2022]
Abstract
"San Huang Powder," a nonsterile milled herb powder, is frequently used to treat burn wounds in traditional Chinese herbal medicine. However, treating a wound with a nonsterile dressing or reagent is not compatible with the current guidelines in modern medicine. Therefore, we investigated the bactericidal and anti-inflammatory activities of four herb extracts used in "San Huang Powder" in vitro. Meanwhile, an in vivo porcine model with superficial second-degree burns was used for the experiments since the size and skin composition of pigs are the closest to that of the human body. The minimal bactericidal concentration (MBC) of the herb extracts was determined. The in vitro assay indicated that Rhubarb and Phellodendron bark extracts decreased the levels of inflammatory cytokines, IL-8, and GM-CSF on LPS-induced HMEC-1 cells. In accordance with this result, the histopathological evaluation results showed that the efficacy of "San Huang Powder" containing both herb materials was much better than the group without Rhubarb. Our results not only provide a basis to understand why "San Huang Powder" has been used to clinically treat wounds without sterilization directly since ancient times but also show the advantages of using multiple herb materials simultaneously on wound sites to prevent infection during treatment. Rhubarb is the recommended ingredient involved in the preparation of "San Huang Powder" to ensure the healing efficacy of burn wounds.
Collapse
|
7
|
Litchfield M, Wuest M, Glubrecht D, Briard E, Auberson YP, McMullen TPW, Brindley DN, Wuest F. Positron Emission Tomography Imaging of Autotaxin in Thyroid and Breast Cancer Models Using [ 18F]PRIMATX. Mol Pharm 2021; 18:3352-3364. [PMID: 34319110 DOI: 10.1021/acs.molpharmaceut.1c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Autotaxin (ATX) is a secreted enzyme responsible for producing lysophosphatidic acid (LPA). The ATX/LPA signaling axis is typically activated in wound healing and tissue repair processes. The ATX/LPA axis is highjacked and upregulated in the progression and persistence of several chronic inflammatory diseases, including cancer. As ATX inhibitors are now progressing to clinical testing, innovative diagnostic tools such as positron emission tomography (PET) are needed to measure ATX expression in vivo accurately. The radiotracer, [18F]PRIMATX, was recently developed and tested for PET imaging of ATX in vivo in a murine melanoma model. The goal of the present work was to further validate [18F]PRIMATX as a PET imaging agent by analyzing its in vivo metabolic stability and suitability for PET imaging of ATX in models of human 8305C thyroid tumor and murine 4T1 breast cancer. [18F]PRIMATX displayed favorable metabolic stability in vivo (65% of intact radiotracer after 60 min p.i.) and provided sufficient tumor uptake profiles in both tumor models. Radiotracer uptake could be blocked by 8-12% in 8305C thyroid tumors in the presence of ATX inhibitor AE-32-NZ70 as determined by PET and ex vivo biodistribution analyses. [18F]PRIMATX also showed high brain uptake, which was reduced by 50% through the administration of ATX inhibitor AE-32-NZ70. [18F]PRIMATX is a suitable radiotracer for PET imaging of ATX in the brain and peripheral tumor tissues.
Collapse
Affiliation(s)
- Marcus Litchfield
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Alberta, Canada
| | - Melinda Wuest
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada
| | - Daryl Glubrecht
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada
| | - Emmanuelle Briard
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabristrasse 2, Novartis Campus, Basel CH-4056, Switzerland
| | - Yves P Auberson
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabristrasse 2, Novartis Campus, Basel CH-4056, Switzerland
| | - Todd P W McMullen
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Alberta, Canada
| | - David N Brindley
- Department of Biochemistry, University of Alberta, Edmonton T6G 1Z2, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Alberta, Canada
| | - Frank Wuest
- Department of Oncology, University of Alberta, 11560 University Avenue, Edmonton T6G 1Z2, Alberta, Canada.,Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2S2, Alberta, Canada
| |
Collapse
|
8
|
Huo Z, Lomora M, Kym U, Palivan C, Holland-Cunz SG, Gros SJ. AQP1 Is Up-Regulated by Hypoxia and Leads to Increased Cell Water Permeability, Motility, and Migration in Neuroblastoma. Front Cell Dev Biol 2021; 9:605272. [PMID: 33644043 PMCID: PMC7905035 DOI: 10.3389/fcell.2021.605272] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
The water channel aquaporin 1 (AQP1) has been implicated in tumor progression and metastasis. It is hypothesized that AQP1 expression can facilitate the transmembrane water transport leading to changes in cell structure that promote migration. Its impact in neuroblastoma has not been addressed so far. The objectives of this study have been to determine whether AQP1 expression in neuroblastoma is dependent on hypoxia, to demonstrate whether AQP1 is functionally relevant for migration, and to further define AQP1-dependent properties of the migrating cells. This was determined by investigating the reaction of neuroblastoma cell lines, particularly SH-SY5Y, Kelly, SH-EP Tet-21/N and SK-N-BE(2)-M17 to hypoxia, quantitating the AQP1-related water permeability by stopped-flow spectroscopy, and studying the migration-related properties of the cells in a modified transwell assay. We find that AQP1 expression in neuroblastoma cells is up-regulated by hypoxic conditions, and that increased AQP1 expression enabled the cells to form a phenotype which is associated with migratory properties and increased cell agility. This suggests that the hypoxic tumor microenvironment is the trigger for some tumor cells to transition to a migratory phenotype. We demonstrate that migrating tumor cell express elevated AQP1 levels and a hypoxic biochemical phenotype. Our experiments strongly suggest that elevated AQP1 might be a key driver in transitioning stable tumor cells to migrating tumor cells in a hypoxic microenvironment.
Collapse
Affiliation(s)
- Zihe Huo
- Department of Pediatric Surgery, University Children's Hospital Basel, Basel, Switzerland.,Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Mihai Lomora
- Department of Physical Chemistry, University of Basel, Basel, Switzerland
| | - Urs Kym
- Department of Pediatric Surgery, University Children's Hospital Basel, Basel, Switzerland.,Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Cornelia Palivan
- Department of Physical Chemistry, University of Basel, Basel, Switzerland
| | - Stefan G Holland-Cunz
- Department of Pediatric Surgery, University Children's Hospital Basel, Basel, Switzerland.,Department of Clinical Research, University of Basel, Basel, Switzerland
| | - Stephanie J Gros
- Department of Pediatric Surgery, University Children's Hospital Basel, Basel, Switzerland.,Department of Clinical Research, University of Basel, Basel, Switzerland
| |
Collapse
|
9
|
D'Aprile C, Prioni S, Mauri L, Prinetti A, Grassi S. Lipid rafts as platforms for sphingosine 1-phosphate metabolism and signalling. Cell Signal 2021; 80:109929. [PMID: 33493577 DOI: 10.1016/j.cellsig.2021.109929] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Spontaneous segregation of cholesterol and sphingolipids as a liquid-ordered phase leads to their clustering in selected membrane areas, the lipid rafts. These specialized membrane domains enriched in gangliosides, sphingomyelin, cholesterol and selected proteins involved in signal transduction, organize and determine the function of multiprotein complexes involved in several aspects of signal transduction, thus regulating cell homeostasis. Sphingosine 1-phosphate, an important biologically active mediator, is involved in several signal transduction processes regulating a plethora of cell functions and, not only several of its downstream effectors tend to localize in lipid rafts, some of the enzymes involved in its pathway, of receptors involved in its signalling and its transporters have been often found in these membrane microdomains. Considering this, in this review we address what is currently known regarding the relationship between sphingosine 1-phosphate metabolism and signalling and plasma membrane lipid rafts.
Collapse
Affiliation(s)
- Chiara D'Aprile
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| |
Collapse
|
10
|
Pini N, Huo Z, Kym U, Holland-Cunz S, Gros SJ. AQP1-Driven Migration Is Independent of Other Known Adverse Factors but Requires a Hypoxic Undifferentiated Cell Profile in Neuroblastoma. CHILDREN-BASEL 2021; 8:children8010048. [PMID: 33467498 PMCID: PMC7829990 DOI: 10.3390/children8010048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/30/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
Neuroblastoma is a biologically very heterogeneous tumor with its clinical manifestation ranging from spontaneous regression to highly aggressive metastatic disease. Several adverse factors have been linked to oncogenesis, tumor progression and metastases of neuroblastoma including NMYC amplification, the neural adhesion molecule NCAM, as well as CXCR4 as a promoter of metastases. In this study, we investigate to what extent the expression of AQP1 in neuroblastoma correlates with changing cellular factors such as the hypoxic status, differentiation, expression of known adverse factors such as NMYC and NCAM, and CXCR4-related metastatic spread. Our results show that while AQP1 expression leads to an increased migratory behavior of neuroblastoma cells under hypoxic conditions, we find that hypoxia is associated with a reduction of NMYC in the same cells. A similar effect can be observed when using the tetracycline driven mechanism of SH-EP/Tet cells. When NMYC is not expressed, the expression of AQP1 is increased together with an increased expression of HIF-1α and HIF-2α. We furthermore show that when growing cells in different cell densities, they express AQP1, HIF-1α, HIF-2α, NMYC and NCAM to different degrees. AQP1 expression correlates with a hypoxic profile of these cells with increased HIF-1α and HIF-2α expression, as well as with NMYC and NCAM expression in two out of three neuroblastoma cell lines. When investigating cell properties of the cells that actually migrate, we find that the increased APQ1 expression in the migrated cells correlates with an increased NMYC and NCAM expression again in two out of three cell lines. Expression of the tumor cell homing marker CXCR4 varies between different tumor areas and between cell lines. While some migrated tumor cells highly express CXCR4, cells of other origin do not. In the initial phase of migration, we determined a dominant role of AQP1 expression of migrating cells in the scratch assay.
Collapse
Affiliation(s)
- Nicola Pini
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (N.P.); (Z.H.); (U.K.); (S.H.-C.)
| | - Zihe Huo
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (N.P.); (Z.H.); (U.K.); (S.H.-C.)
| | - Urs Kym
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (N.P.); (Z.H.); (U.K.); (S.H.-C.)
| | - Stefan Holland-Cunz
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (N.P.); (Z.H.); (U.K.); (S.H.-C.)
| | - Stephanie J. Gros
- Department of Pediatric Surgery, University Children’s Hospital Basel, 4031 Basel, Switzerland; (N.P.); (Z.H.); (U.K.); (S.H.-C.)
- Department of Clinical Research, University of Basel, 4031 Basel, Switzerland
- Correspondence:
| |
Collapse
|
11
|
Emerging roles of lysophospholipids in health and disease. Prog Lipid Res 2020; 80:101068. [PMID: 33068601 DOI: 10.1016/j.plipres.2020.101068] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/09/2020] [Accepted: 10/11/2020] [Indexed: 12/22/2022]
Abstract
Lipids are abundant and play essential roles in human health and disease. The main functions of lipids are building blocks for membrane biogenesis. However, lipids are also metabolized to produce signaling molecules. Here, we discuss the emerging roles of circulating lysophospholipids. These lysophospholipids consist of lysoglycerophospholipids and lysosphingolipids. They are both present in cells at low concentration, but their concentrations in extracellular fluids are significantly higher. The biological functions of some of these lysophospholipids have been recently revealed. Remarkably, some of the lysophospholipids play pivotal signaling roles as well as being precursors for membrane biogenesis. Revealing how circulating lysophospholipids are produced, released, transported, and utilized in multi-organ systems is critical to understand their functions. The discovery of enzymes, carriers, transporters, and membrane receptors for these lysophospholipids has shed light on their physiological significance. In this review, we summarize the biological roles of these lysophospholipids via discussing about the proteins regulating their functions. We also discuss about their potential impacts to human health and diseases.
Collapse
|
12
|
Lee D, Kim YH, Kim JH. The Role of Lysophosphatidic Acid in Adult Stem Cells. Int J Stem Cells 2020; 13:182-191. [PMID: 32587135 PMCID: PMC7378901 DOI: 10.15283/ijsc20035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/24/2020] [Accepted: 05/21/2020] [Indexed: 01/06/2023] Open
Abstract
Stem cells are undifferentiated multipotent precursor cells that are capable both of perpetuating themselves as stem cells (self-renewal) and of undergoing differentiation into one or more specialized types of cells. And these stem cells have been reported to reside within distinct anatomic locations termed “niches”. The long-term goals of stem cell biology range from an understanding of cell-lineage determination and tissue organization to cellular therapeutics for degenerative diseases. Stem cells maintain tissue function throughout an organism’s lifespan by replacing differentiated cells. To perform this function, stem cells provide a unique combination of multilineage developmental potential and the capacity to undergo self-renewing divisions. The loss of self-renewal capacity in stem cells underlies certain degenerative diseases and the aging process. This self-renewal regulation must balance the regenerative needs of tissues that persist throughout life. Recent evidence suggests lysophosphatidic acid (LPA) signaling pathway plays an important role in the regulation of a variety of stem cells. In this review, we summarize the evidence linking between LPA and stem cell regulation. The LPA-induced signaling pathway regulates the proliferation and survival of stem cells and progenitors, and thus are likely to play a role in the maintenance of stem cell population in the body. This lipid mediator regulatory system can be a novel potential therapeutics for stem cell maintenance.
Collapse
Affiliation(s)
- Dongjun Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Korea
| | - Yun Hak Kim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Korea.,Department of Biomedical Informatics, Pusan National University School of Medicine, Yangsan, Korea
| | - Jae Ho Kim
- Department of Physiology, Pusan National University School of Medicine, Yangsan, Korea
| |
Collapse
|
13
|
Bone regenerative potential of the selective sphingosine 1-phosphate receptor modulator siponimod: In vitro characterisation using osteoblast and endothelial cells. Eur J Pharmacol 2020; 882:173262. [PMID: 32534075 DOI: 10.1016/j.ejphar.2020.173262] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 12/31/2022]
Abstract
The repair of critical bone defects remains a significant therapeutic challenge. While the implantation of drug-eluting scaffolds is an option, a drug with the optimal pharmacological properties has not yet been identified. Agents acting at sphingosine 1-phosphate (S1P) receptors have been considered, but those investigated so far do not discriminate between the five known S1P receptors. This work was undertaken to investigate the potential of the specific S1P1/5 modulator siponimod as a bone regenerative agent, by testing in vitro its effect on cell types critical to the bone regeneration process. hFOB osteoblasts and HUVEC endothelial cells were treated with siponimod and other S1P receptor modulators and investigated for changes in intracellular cyclic AMP content, viability, proliferation, differentiation, attachment and cellular motility. Siponimod showed no effect on the viability and proliferation of osteoblasts and endothelial cells, but increased osteoblast differentiation (as shown by increased alkaline phosphatase activity). Furthermore, siponimod significantly increased endothelial cell motility in scratch and transwell migration assays. These effects on osteoblast differentiation and endothelial cell migration suggest that siponimod may be a potential agent for the stimulation of localised differentiation of osteoblasts in critical bone defects.
Collapse
|
14
|
Platelet and hemoglobin count at diagnosis are associated with survival in African American and Caucasian patients with colorectal cancer. Cancer Epidemiol 2020; 67:101746. [PMID: 32521488 DOI: 10.1016/j.canep.2020.101746] [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: 02/04/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND African Americans (AAs) compared to Caucasian Americans (CAs) with colorectal cancer (CRC) have lower stage-specific survival. CRC patients often present with several hematopathologies (such as thrombocytosis, thrombocytopenia, anemia) at diagnosis, which is associated with poorer survival. However, whether these measures impact the racial disparity in survival is not known. METHODS The study population was composed of 581 histologically confirmed CRCs at the Medical University of South Carolina (393 CA, 188 AA) diagnosed between 01/01/2000 and 06/30/2013. We used Cox proportional hazards regression to estimate the association between thrombocytosis, thrombocytopenia, or anemia at diagnosis and risk of death by race. This analysis was adjusted for age, sex, stage and first-line treatment. RESULTS In all patients combined, thrombocytosis, thrombocytopenia, and anemia (vs. the normal ranges) were associated with significantly higher risks of death. In the race-specific analyses, AAs (HR 2.51 [95 % CI: 1.52-4.15]) vs. CAs (HR 1.15 [95 % CI: 0.75-1.75]) with thrombocytosis compared to normal had a higher risk of death (p for difference = 0.03). CONCLUSIONS Abnormal thrombocyte and hemoglobin levels at diagnosis were associated with poorer survival. AAs compared to CAs with elevated platelets at diagnosis had a higher risk of death. Our study is the first to examine the role of race, hematologic measures at diagnosis, and risk of death in colorectal cancer patients. These results suggest that the racial differences in the immune response may contribute to the racial disparity in survival.
Collapse
|
15
|
Wu XN, Ma YY, Hao ZC, Wang H. [Research progress on the biological regulatory function of lysophosphatidic acid in bone tissue cells]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2020; 38:324-329. [PMID: 32573143 DOI: 10.7518/hxkq.2020.03.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Lysophosphatidic acid (LPA) is a small phospholipid that is present in all eukaryotic tissues and blood plasma. As an extracellular signaling molecule, LPA mediates many cellular functions by binding to six known G protein-coupled receptors and activating their downstream signaling pathways. These functions indicate that LPA may play important roles in many biological processes that include organismal development, wound healing, and carcinogenesis. Recently, many studies have found that LPA has various biological effects in different kinds of bone cells. These findings suggest that LPA is a potent regulator of bone development and remodeling and holds promising application potential in bone tissue engineering. Here, we review the recent progress on the biological regulatory function of LPA in bone tissue cells.
Collapse
Affiliation(s)
- Xiang-Nan Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China;Hospital of Stomatology, Sun Yat-sen University, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Yuan-Yuan Ma
- Hospital of Stomatology, Sun Yat-sen University, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Zhi-Chao Hao
- Hospital of Stomatology, Sun Yat-sen University, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Hang Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
16
|
Rothe R, Schulze S, Neuber C, Hauser S, Rammelt S, Pietzsch J. Adjuvant drug-assisted bone healing: Part II - Modulation of angiogenesis. Clin Hemorheol Microcirc 2020; 73:409-438. [PMID: 31177206 DOI: 10.3233/ch-199103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The treatment of critical-size bone defects following complicated fractures, infections or tumor resections is a major challenge. The same applies to fractures in patients with impaired bone healing due to systemic inflammatory and metabolic diseases. Despite considerable progress in development and establishment of new surgical techniques, design of bone graft substitutes and imaging techniques, these scenarios still represent unresolved clinical problems. However, the development of new active substances offers novel potential solutions for these issues. This work discusses therapeutic approaches that influence angiogenesis or hypoxic situations in healing bone and surrounding tissue. In particular, literature on sphingosine-1-phosphate receptor modulators and nitric oxide (NO•) donors, including bi-functional (hybrid) compounds like NO•-releasing cyclooxygenase-2 inhibitors, was critically reviewed with regard to their local and systemic mode of action.
Collapse
Affiliation(s)
- Rebecca Rothe
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sabine Schulze
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Stefan Rammelt
- University Center of Orthopaedics and Traumatology (OUC), University Hospital Carl Gustav Carus, Dresden, Germany.,Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), Tatzberg 4, Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Dresden, Germany
| |
Collapse
|
17
|
Mir SS, Bhat HF, Bhat ZF. Dynamic actin remodeling in response to lysophosphatidic acid. J Biomol Struct Dyn 2020; 38:5253-5265. [PMID: 31920158 DOI: 10.1080/07391102.2019.1696230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Lysophosphatidic acid (LPA) is a multifunctional regulator of actin cytoskeleton that exerts a dramatic impact on the actin cytoskeleton to build a platform for diverse cellular processes including growth cone guidance, neurite retraction and cell motility. It has been implicated in the formation and dissociation of complexes between actin and actin binding proteins, supporting its role in actin remodeling. Several studies point towards its ability to facilitate formation of special cellular structures including focal adhesions and actin stress fibres by phosphoregulation of several actin associated proteins and their multiple regulatory kinases and phosphatases. In addition, multiple levels of crosstalk among the signaling cascades activated by LPA, affect actin cytoskeleton-mediated cell migration and chemotaxis which in turn play a crucial role in cancer metastasis. In the current review, we have attempted to highlight the role of LPA as an actin modulator which functions by controlling activities of specific cellular proteins that underlie mechanisms employed in cytoskeletal and pathophysiological events within the cell. Further studies on the actin affecting/remodeling activity of LPA in different cell types will no doubt throw up many surprises essential to gain a full understanding of its contribution in physiological processes as well as in diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saima S Mir
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu And Kashmir, India.,Division of Animal Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, Jammu And Kashmir, India
| | - Hina F Bhat
- Division of Animal Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Srinagar, Jammu And Kashmir, India
| | - Zuhaib F Bhat
- Department of Wine, Food & Molecular Biosciences, Lincoln University, Lincoln, New Zealand.,Division of Livestock Products and Technology, Sher-e-Kashmir University of Agricultural Sciences and Technology of Jammu (SKUAST-J), R.S. Pora, Jammu And Kashmir, India
| |
Collapse
|
18
|
Lin CH, Lu JH, Hsia K, Lee H, Yao CL, Ma H. The Antithrombotic Function of Sphingosine-1-Phosphate on Human Adipose-Stem-Cell-Recellularized Tissue Engineered Vascular Graft In Vitro. Int J Mol Sci 2019; 20:ijms20205218. [PMID: 31640220 PMCID: PMC6829437 DOI: 10.3390/ijms20205218] [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: 07/26/2019] [Revised: 10/11/2019] [Accepted: 10/17/2019] [Indexed: 12/26/2022] Open
Abstract
Adipose stem cells (ASCs) show potential in the recellularization of tissue engineerined vascular grafts (TEVGs). However, whether sphingosine-1-phosphate (S1P) could further enhance the adhesion, proliferation, and antithrombosis of ASCs on decellularized vascular scaffolds is unknown. This study investigated the effect of S1P on the recellularization of TEVGs with ASCs. Human ASCs were derived from lipoaspirate. Scaffolds were derived from human umbilical arteries (HUAs) with treatment of 0.1% sodium dodecyl sulfate (SDS) for 48 h (decellularized HUAs; DHUAs). The adhesion, proliferation, and antithrombotic functions (kinetic clotting time and platelet adhesion) of ASCs on DHUAs with S1P or without S1P were evaluated. The histology and DNA examination revealed a preserved structure and the elimination of the nuclear component more than 95% in HUAs after decellularizaiton. Human ASCs (hASCs) showed CD29(+), CD73(+), CD90(+), CD105(+), CD31(-), CD34(-), CD44(-), HLA-DR(-), and CD146(-) while S1P-treated ASCs showed marker shifting to CD31(+). In contrast to human umbilical vein endothelial cells (HUVECs), S1P didn't significantly increase proliferation of ASCs on DHUAs. However, the kinetic clotting test revealed prolonged blood clotting in S1P-treated ASC-recellularized DHUAs. S1P also decreased platelet adhesion on ASC-recellularized DHUAs. In addition, S1P treatment increased the syndecan-1 expression of ASCs. TEVG reconstituted with S1P and ASC-recellularized DHUAs showed an antithrombotic effect in vitro. The preliminary results showed that ASCs could adhere to DHUAs and S1P could increase the antithrombotic effect on ASC-recellularized DHUAs. The antithrombotic effect is related to ASCs exhibiting an endothelial-cell-like function and preventing of syndecan-1 shedding. A future animal study is warranted to prove this novel method.
Collapse
Affiliation(s)
- Chih-Hsun Lin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Department of Surgery, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Jen-Her Lu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Department of Surgery, medicine & Pediatrics, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan.
- Department of Pediatrics, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Kai Hsia
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan.
| | - Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan.
| | - Chao-Ling Yao
- Department of Chemical Engineering and Materials Science, Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Chung-Li, Taoyuan City 32003, Taiwan.
| | - Hsu Ma
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- Department of Surgery, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- Department of Surgery, medicine & Pediatrics, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan.
| |
Collapse
|
19
|
Zhao Y, Hasse S, Zhao C, Bourgoin SG. Targeting the autotaxin - Lysophosphatidic acid receptor axis in cardiovascular diseases. Biochem Pharmacol 2019; 164:74-81. [PMID: 30928673 DOI: 10.1016/j.bcp.2019.03.035] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/26/2019] [Indexed: 02/06/2023]
Abstract
Lysophosphatidic acid (LPA) is a well-characterized bioactive lipid mediator, which is involved in development, physiology, and pathological processes of the cardiovascular system. LPA can be produced both inside cells and in biological fluids. The majority of extracellularLPAis produced locally by the secreted lysophospholipase D, autotaxin (ATX), through its binding to various β integrins or heparin sulfate on cell surface and hydrolyzing various lysophospholipids. LPA initiates cellular signalling pathways upon binding to and activation of its G protein-coupled receptors (LPA1-6). LPA has potent effects on various blood cells and vascular cells involved in the development of cardiovascular diseases such as atherosclerosis and aortic valve sclerosis. LPA signalling drives cell migration and proliferation, cytokine production, thrombosis, fibrosis, as well as angiogenesis. For instance, LPA promotes activation and aggregation of platelets through LPA5, increases expression of adhesion molecules in endothelial cells, and enhances expression of tissue factor in vascular smooth muscle cells. Furthermore, LPA induces differentiation of monocytes into macrophages and stimulates oxidized low-density lipoproteins (oxLDLs) uptake by macrophages to form foam cells during formation of atherosclerotic lesions through LPA1-3. This review summarizes recent findings of the roles played by ATX, LPA and LPA receptors (LPARs) in atherosclerosis and calcific aortic valve disease. Targeting the ATX-LPAR axis may have potential applications for treatment of patients suffering from various cardiovascular diseases.
Collapse
Affiliation(s)
- Yang Zhao
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V4G2, Canada
| | - Stephan Hasse
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V4G2, Canada
| | - Chenqi Zhao
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada
| | - Sylvain G Bourgoin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Canada; Département de microbiologie, infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, QC G1V4G2, Canada.
| |
Collapse
|
20
|
Chabowski DS, Kadlec AO, Ait‐Aissa K, Hockenberry JC, Pearson PJ, Beyer AM, Gutterman DD. Lysophosphatidic acid acts on LPA 1 receptor to increase H 2 O 2 during flow-induced dilation in human adipose arterioles. Br J Pharmacol 2018; 175:4266-4280. [PMID: 30153326 PMCID: PMC6193883 DOI: 10.1111/bph.14492] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/23/2018] [Accepted: 08/09/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE NO produces arteriolar flow-induced dilation (FID) in healthy subjects but is replaced by mitochondria-derived hydrogen peroxide (mtH2 O2 ) in patients with coronary artery disease (CAD). Lysophosphatidic acid (LPA) is elevated in patients with risk factors for CAD, but its functional effect in arterioles is unknown. We tested whether elevated LPA changes the mediator of FID from NO to mtH2 O2 in human visceral and subcutaneous adipose arterioles. EXPERIMENTAL APPROACH Arterioles were cannulated on glass micropipettes and pressurized to 60 mmHg. We recorded lumen diameter after graded increases in flow in the presence of either NOS inhibition (L-NAME) or H2 O2 scavenging (Peg-Cat) ± LPA (10 μM, 30 min), ±LPA1 /LPA3 receptor antagonist (Ki16425) or LPA2 receptor antagonist (H2L5186303). We analysed LPA receptor RNA and protein levels in human arterioles and human cultured endothelial cells. KEY RESULTS FID was inhibited by L-NAME but not Peg-Cat in untreated vessels. In vessels treated with LPA, FID was of similar magnitude but inhibited by Peg-Cat while L-NAME had no effect. Rotenone attenuated FID in vessels treated with LPA indicating mitochondria as a source of ROS. RNA transcripts from LPA1 and LPA2 but not LPA3 receptors were detected in arterioles. LPA1 but not LPA3 receptor protein was detected by Western blot. Pretreatment of vessels with an LPA1 /LPA3 , but not LPA2 , receptor antagonist prior to LPA preserved NO-mediated dilation. CONCLUSIONS AND IMPLICATIONS These findings suggest an LPA1 receptor-dependent pathway by which LPA increases arteriolar release of mtH2 O2 as a mediator of FMD.
Collapse
Affiliation(s)
- Dawid S Chabowski
- Department of Pharmacology and ToxicologyMedical College of WisconsinMilwaukeeWIUSA
| | - Andrew O Kadlec
- Department of PhysiologyMedical College of WisconsinMilwaukeeWIUSA
| | - Karima Ait‐Aissa
- Department of Medicine – Cardiovascular CenterMedical College of WisconsinMilwaukeeWIUSA
| | - Joseph C Hockenberry
- Department of Medicine – Cardiovascular CenterMedical College of WisconsinMilwaukeeWIUSA
| | - Paul J Pearson
- Department of Surgery – Cardiothoracic SurgeryMedical College of WisconsinMilwaukeeWIUSA
| | - Andreas M Beyer
- Department of PhysiologyMedical College of WisconsinMilwaukeeWIUSA
- Department of Medicine – Cardiovascular CenterMedical College of WisconsinMilwaukeeWIUSA
| | - David D Gutterman
- Department of Pharmacology and ToxicologyMedical College of WisconsinMilwaukeeWIUSA
- Department of PhysiologyMedical College of WisconsinMilwaukeeWIUSA
- Department of Medicine – Cardiovascular CenterMedical College of WisconsinMilwaukeeWIUSA
- VA Medical CenterMilwaukeeWIUSA
| |
Collapse
|
21
|
Shen X, Zou J, Li F, Zhang T, Guo T. Lysophosphatidic acid enhances neointimal hyperplasia following vascular injury through modulating proliferation, autophagy, inflammation and oxidative stress. Mol Med Rep 2018; 18:87-96. [PMID: 29749484 PMCID: PMC6059717 DOI: 10.3892/mmr.2018.8937] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/21/2018] [Indexed: 01/15/2023] Open
Abstract
Lysophosphatidic acid (LPA), which is one of the intermediate products of membrane phospholipid metabolism, is a bioactive phospholipid that possesses diverse activities. In the present study, the effects of LPA on neointimal formation following vascular injury were investigated. A carotid artery balloon injury model was employed in the present study, and following vascular injury, rats received an intraperitoneal injection of 1 mg/kg LPA. Subsequently, histopathological alterations were assessed by hematoxylin and eosin staining, the expression levels of proliferating cell nuclear antigen (PCNA) were detected by immunohistochemistry, apoptosis was assessed via a terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, and the expression levels of apoptosis-associated and autophagy-associated proteins were detected by western blotting. In addition, inflammatory and oxidative stress-associated factors were assessed by reverse transcription-quantitative polymerase chain reaction or corresponding kits. The results of the present study demonstrated that LPA enhanced vascular injury-induced neointimal hyperplasia. LPA further elevated the expression levels of PCNA in the injured carotid artery tissues. LPA exhibited no effect on apoptosis in carotid artery tissues, whereas it modulated autophagy in the injured carotid artery tissues. Furthermore, LPA enhanced vascular injury-induced inflammation and oxidative stress. The present study demonstrated that LPA may enhance neointimal hyperplasia following vascular injury by modulating proliferation, autophagy, inflammation and oxidative stress, but not apoptosis. Furthermore LPA may contribute to the pathology of atherosclerosis and may be considered a promising therapeutic target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Xuhui Shen
- Third Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, P.R. China
| | - Jianjun Zou
- Third Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, P.R. China
| | - Fuyong Li
- Third Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, P.R. China
| | - Tianhe Zhang
- Third Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, P.R. China
| | - Tongqi Guo
- Third Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
22
|
Dorotea D, Cho A, Lee G, Kwon G, Lee J, Sahu PK, Jeong LS, Cha DR, Ha H. Orally active, species-independent novel A 3 adenosine receptor antagonist protects against kidney injury in db/db mice. Exp Mol Med 2018; 50:1-14. [PMID: 29674631 PMCID: PMC5938017 DOI: 10.1038/s12276-018-0053-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 11/25/2017] [Accepted: 12/18/2017] [Indexed: 11/22/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage kidney disease, and the current pharmacological treatment for DKD is limited to renin-angiotensin system (RAS) inhibitors. Adenosine is detectable in the kidney and is significantly elevated in response to cellular damage. While all 4 known subtypes of adenosine receptors, namely, A1AR, A2aAR, A2bAR, and A3AR, are expressed in the kidney, our previous study has demonstrated that a novel, orally active, species-independent, and selective A3AR antagonist, LJ-1888, ameliorates unilateral ureteral obstruction-induced tubulointerstitial fibrosis. The present study examined the protective effects of LJ-2698, which has higher affinity and selectivity for A3AR than LJ-1888, on DKD. In experiment I, dose-dependent effects of LJ-2698 were examined by orally administering 1.5, 5, or 10 mg/kg for 12 weeks to 8-week-old db/db mice. In experiment II, the effects of LJ-2698 (10 mg/kg) were compared to those of losartan (1.5 mg/kg), which is a standard treatment for patients with DKD. LJ-2698 effectively prevented kidney injuries such as albuminuria, glomerular hypertrophy, tubular injury, podocyte injury, fibrosis, inflammation, and oxidative stress in diabetic mice as much as losartan. In addition, inhibition of lipid accumulation along with increases in PGC1α, a master regulator of mitochondrial biogenesis, were demonstrated in diabetic mice treated with either LJ-2698 or losartan. These results suggest that LJ-2698, a selective A3AR antagonist, may become a novel therapeutic agent against DKD. A therapeutic treatment targeting a protein involved in the progression of diabetic kidney disease (DKD) shows promise in mouse trials. Between 30 and 40 per cent of diabetic patients suffer from DKD, a common cause to fatal end-stage kidney disease. Protein receptors, commonly expressed on cell surfaces throughout the body, play both positive and negative roles in diseases. The A3 adenosine receptor (A3AR) is highly expressed in diabetic kidney tissue, and is linked to disease progression. Hunjoo Ha at Ewha Womans University in Seoul, Republic of Korea, and co-workers demonstrated the positive effect of a novel drug in targeting A3AR in mice with DKD. A 12-week treatment of the drug prevented kidney injury, lowered oxidative stress and inflammation, and improved kidney function. It may prove an invaluable drug, particularly in combination with an existing DKD drug.
Collapse
Affiliation(s)
- Debra Dorotea
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Ahreum Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Gayoung Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Guideock Kwon
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Junghwa Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Pramod K Sahu
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, Korea.,Future Medicine Co, Seoul, Korea
| | - Lak Shin Jeong
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Dae Ryong Cha
- Department of Nephrology, Korea University Ansan Hospital, Ansan, Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea.
| |
Collapse
|
23
|
Induction of hemangiosarcoma in mice after chronic treatment with S1P-modulator siponimod and its lack of relevance to rat and human. Arch Toxicol 2018; 92:1877-1891. [PMID: 29556671 PMCID: PMC5962627 DOI: 10.1007/s00204-018-2189-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/13/2018] [Indexed: 11/30/2022]
Abstract
A high incidence of hemangiosarcoma (HSA) was observed in mice treated for 2 years with siponimod, a sphingosine-1-phosphate receptor 1 (S1P1) functional antagonist, while no such tumors were observed in rats under the same treatment conditions. In 3-month rat (90 mg/kg/day) and 9-month mouse (25 and 75 mg/kg/day) in vivo mechanistic studies, vascular endothelial cell (VEC) activation was observed in both species, but VEC proliferation and persistent increases in circulating placental growth factor 2 (PLGF2) were only seen in the mouse. In mice, these effects were sustained over the 9-month study duration, while in rats increased mitotic gene expression was present at day 3 only and PLGF2 was induced only during the first week of treatment. In the mouse, the persistent VEC activation, mitosis induction, and PLGF2 stimulation likely led to sustained neo-angiogenesis which over life-long treatment may result in HSA formation. In rats, despite sustained VEC activation, the transient mitotic and PLGF2 stimuli did not result in the formation of HSA. In vitro, the mouse and rat primary endothelial cell cultures mirrored their respective in vivo findings for cell proliferation and PLGF2 release. Human VECs, like rat cells, were unresponsive to siponimod treatment with no proliferative response and no release of PLGF2 at all tested concentrations. Hence, it is suggested that the human cells also reproduce a lack of in vivo response to siponimod. In conclusion, the molecular mechanisms leading to siponimod-induced HSA in mice are considered species specific and likely irrelevant to humans.
Collapse
|
24
|
Lan T, Hung SH, Su X, Wong SWK, Tseng Y. Integrating transient cellular and nuclear motions to comprehensively describe cell migration patterns. Sci Rep 2018; 8:1488. [PMID: 29367613 PMCID: PMC5784082 DOI: 10.1038/s41598-018-19885-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 01/09/2018] [Indexed: 02/07/2023] Open
Abstract
Various subcellular activities, such as protrusion and detachment, compose a cell migration process. The molecular mechanisms of these subcellular activities have been elucidated. However, there is no method that can assess the contributions of these subcellular activities to the global cell migration pattern of a given cell type. Hence, we develop a powerful approach based on CN correlations that quantitatively profiles the cell migration pattern of a given cell type in terms of assembled subcellular activities. In this way, we bridge migration data at the cellular level with underlying molecular mechanisms. The CN correlation profile is found to uniquely and consistently represent the cell migration pattern of each cell type probed. It can clearly reveal the effects of molecular perturbations, such as Y27632 and Cdc42 knockdown on each subcellular migratory activity. As a result, the CN correlation approach serves as a cell dynamic descriptor that can extract comprehensive quantitative data from cell migration movies for integrative biological analyses.
Collapse
Affiliation(s)
- Tian Lan
- Department of Chemical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Shen-Hsiu Hung
- Department of Chemical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Xudong Su
- Department of Chemical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Samuel W K Wong
- Department of Statistics, University of Florida, Gainesville, FL, 32611, USA
| | - Yiider Tseng
- Department of Chemical Engineering, University of Florida, Gainesville, FL, 32611, USA. .,J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA. .,Institute for Cell & Tissue Science and Engineering, University of Florida, Gainesville, FL, 32611, USA. .,National Cancer Institute-Physical Science Oncology Center, Gainesville, FL, 32611, USA.
| |
Collapse
|
25
|
Yang W, Deng Y, Zhou H, Jiang H, Li Y, Chu Y, Wang X, Gong L. Metabolic characteristics of Rhizoma Coptidis intervention in spontaneously hypertensive rats: Insights gained from metabolomics analysis of serum. Mol Med Rep 2017; 16:4301-4308. [PMID: 28765928 DOI: 10.3892/mmr.2017.7119] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 05/18/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the intervention mechanisms of Rhizoma Coptidis using spontaneously hypertensive rats. A serum metabolomics analysis was performed with high performance liquid chromatography‑quadrupole/time of flight mass spectrometer in positive mode. The obtained data were further analyzed by principal component and partial least‑squares discriminant analysis to reveal differentiating metabolites. The pattern of metabolites in the serum after Rhizoma Coptidis exhibited distinct alterations. A total of 10 potential biomarkers were significantly altered in the serum and may be associated with the underlying mechanism. Alterations were primarily associated with phospholipid metabolism, fatty acid metabolism, amino acid metabolism and arachidonic acid metabolism. In addition, biochemical alterations in potential biomarkers were associated with inflammation, nitric oxide production, platelet aggregation and endothelial function. By analyzing and verifying the specific biomarkers, metabolomics may be helpful to further understand the underlying therapeutic mechanism of Rhizoma Coptidis. Metabolomics is a powerful tool used to investigate the therapeutic effects of herbal medicine with multiple targets.
Collapse
Affiliation(s)
- Wenqing Yang
- Experimental Center of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Yue Deng
- College of Pharmacy of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Honglei Zhou
- College of Pharmacy of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Haiqiang Jiang
- Experimental Center of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Yunlun Li
- Traditional Chinese Medicine Clinical Research Base for Hypertension, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250011, P.R. China
| | - Yangjun Chu
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Xiaoming Wang
- College of Pharmacy of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Lili Gong
- Experimental Center of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| |
Collapse
|
26
|
Hsia K, Yang MJ, Chen WM, Yao CL, Lin CH, Loong CC, Huang YL, Lin YT, Lander AD, Lee H, Lu JH. Sphingosine-1-phosphate improves endothelialization with reduction of thrombosis in recellularized human umbilical vein graft by inhibiting syndecan-1 shedding in vitro. Acta Biomater 2017; 51:341-350. [PMID: 28110073 DOI: 10.1016/j.actbio.2017.01.050] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 12/31/2022]
Abstract
Sphingosine-1-phosphate (S1P) has been known to promote endothelial cell (EC) proliferation and protect Syndecan-1 (SDC1) from shedding, thereby maintaining this antithrombotic signal. In the present study, we investigated the effect of S1P in the construction of a functional tissue-engineered blood vessel by using human endothelial cells and decellularized human umbilical vein (DHUV) scaffolds. Both human umbilical vein endothelial cells (HUVEC) and human cord blood derived endothelial progenitor cells (EPC) were seeded onto the scaffold with or without the S1P treatment. The efficacy of re-cellularization was determined by using the fluorescent marker CellTracker CMFDA and anti-CD31 immunostaining. The antithrombotic effect of S1P was examined by the anti-aggregation tests measuring platelet adherence and clotting time. Finally, we altered the expression of SDC1, a major glycocalyx protein on the endothelial cell surface, using MMP-7 digestion to explore its role using platelet adhesion tests in vitro. The result showed that S1P enhanced the attachment of HUVEC and EPC. Based on the anti-aggregation tests, S1P-treated HUVEC recellularized vessels when grafted showed reduced thrombus formation compared to controls. Our results also identified reduced SDC1 shedding from HUVEC responsible for inhibition of platelet adherence. However, no significant antithrombogenic effect of S1P was observed on EPC. In conclusion, S1P is an effective agent capable of decreasing thrombotic risk in engineered blood vessel grafts. STATEMENT OF SIGNIFICANCE Sphingosine-1phosphate (S1P) is a low molecular-weight phospholipid mediator that regulates diverse biological activities of endothelial cell, including survival, proliferation, cell barrier integrity, and also influences the development of the vascular system. Based on these characters, we the first time to use it as an additive during the process of a small caliber blood vessel construction by decellularized human umbilical vein and endothelial cell/endothelial progenitor. We further explored the function and mechanism of S1P in promoting revascularization and protection against thrombosis in this tissue engineered vascular grafts. The results showed that S1P could not only accelerate the generation but also reduce thrombus formation of small caliber blood vessel.
Collapse
|
27
|
Priyadarsini S, McKay TB, Sarker-Nag A, Allegood J, Chalfant C, Ma JX, Karamichos D. Complete metabolome and lipidome analysis reveals novel biomarkers in the human diabetic corneal stroma. Exp Eye Res 2016; 153:90-100. [PMID: 27742548 DOI: 10.1016/j.exer.2016.10.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 10/03/2016] [Accepted: 10/07/2016] [Indexed: 01/05/2023]
Abstract
Prolonged hyperglycemia during diabetes mellitus can cause severe ophthalmic complications affecting both the anterior and posterior ocular segments leading to impaired vision or blindness. Diabetes-induced corneal pathologies are associated with decreased wound healing capacity, corneal edema, and altered epithelial basement membrane. The mechanism by which diabetes modulates structure and function within the corneal stroma are unknown. In our study, we characterized the effects of diabetes on extracellular matrix, lipid transport, and cellular metabolism by defining the entire metabolome and lipidome of Type 1 and Type 2 human diabetic corneal stroma. Significant increases in Collagen I and III were found in diabetic corneas suggesting that diabetes promotes defects in matrix structure leading to scarring. Furthermore, increased lipid content, including sphingosine-1-phosphate and dihydrosphingosine, in diabetic corneas compared to healthy controls were measured suggesting altered lipid retention. Metabolomics analysis identified elevated tryptophan metabolites, independent of glucose metabolism, which correlated with upregulation of the Kynurenine pathway in diabetic corneas. We also found significant upregulation of novel biomarkers aminoadipic acid, D,L-pipecolic acid, and dihydroorotate. Our study links aberrant tryptophan metabolism to end-stage pathologies associated with diabetes indicating the potential of the Kynurenine pathway as a therapeutic target for inhibiting diabetes-associated defects in the eye.
Collapse
Affiliation(s)
- Shrestha Priyadarsini
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Tina B McKay
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Akhee Sarker-Nag
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Jeremy Allegood
- Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, VA, USA; Department of Biochemistry and Molecular Biology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; The VCU Johnson Center, Richmond, VA, USA; The VCU Massey Cancer Center, Richmond, VA, USA
| | - Charles Chalfant
- Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, VA, USA; Department of Biochemistry and Molecular Biology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA; The VCU Johnson Center, Richmond, VA, USA; The VCU Massey Cancer Center, Richmond, VA, USA
| | - Jian-Xing Ma
- Department of Physiology Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Dimitrios Karamichos
- Department of Ophthalmology/Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
28
|
Sharma S, Ruffenach G, Umar S, Motayagheni N, Reddy ST, Eghbali M. Role of oxidized lipids in pulmonary arterial hypertension. Pulm Circ 2016; 6:261-73. [PMID: 27683603 DOI: 10.1086/687293] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a multifactorial disease characterized by interplay of many cellular, molecular, and genetic events that lead to excessive proliferation of pulmonary cells, including smooth muscle and endothelial cells; inflammation; and extracellular matrix remodeling. Abnormal vascular changes and structural remodeling associated with PAH culminate in vasoconstriction and obstruction of pulmonary arteries, contributing to increased pulmonary vascular resistance, pulmonary hypertension, and right ventricular failure. The complex molecular mechanisms involved in the pathobiology of PAH are the limiting factors in the development of potential therapeutic interventions for PAH. Over the years, our group and others have demonstrated the critical implication of lipids in the pathogenesis of PAH. This review specifically focuses on the current understanding of the role of oxidized lipids, lipid metabolism, peroxidation, and oxidative stress in the progression of PAH. This review also discusses the relevance of apolipoprotein A-I mimetic peptides and microRNA-193, which are known to regulate the levels of oxidized lipids, as potential therapeutics in PAH.
Collapse
Affiliation(s)
- Salil Sharma
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Grégoire Ruffenach
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Soban Umar
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Negar Motayagheni
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Mansoureh Eghbali
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
29
|
Lan T, Cheng K, Ren T, Arce SH, Tseng Y. Displacement correlations between a single mesenchymal-like cell and its nucleus effectively link subcellular activities and motility in cell migration analysis. Sci Rep 2016; 6:34047. [PMID: 27670131 PMCID: PMC5037420 DOI: 10.1038/srep34047] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 09/07/2016] [Indexed: 01/13/2023] Open
Abstract
Cell migration is an essential process in organism development and physiological maintenance. Although current methods permit accurate comparisons of the effects of molecular manipulations and drug applications on cell motility, effects of alterations in subcellular activities on motility cannot be fully elucidated from those methods. Here, we develop a strategy termed cell-nuclear (CN) correlation to parameterize represented dynamic subcellular activities and to quantify their contributions in mesenchymal-like migration. Based on the biophysical meaning of the CN correlation, we propose a cell migration potential index (CMPI) to measure cell motility. When the effectiveness of CMPI was evaluated with respect to one of the most popular cell migration analysis methods, Persistent Random Walk, we found that the cell motility estimates among six cell lines used in this study were highly consistent between these two approaches. Further evaluations indicated that CMPI can be determined using a shorter time period and smaller cell sample size, and it possesses excellent reliability and applicability, even in the presence of a wide range of noise, as might be generated from individual imaging acquisition systems. The novel approach outlined here introduces a robust strategy through an analysis of subcellular locomotion activities for single cell migration assessment.
Collapse
Affiliation(s)
- Tian Lan
- Department of Chemical Engineering, Gainesville, FL 32611, USA
| | - Kai Cheng
- Department of Chemical Engineering, Gainesville, FL 32611, USA
| | - Tina Ren
- Harvard School of Dental Medicine, Boston, MA 02115, USA
| | | | - Yiider Tseng
- Department of Chemical Engineering, Gainesville, FL 32611, USA.,J. Crayton Pruitt Family Department of Biomedical Engineering, Gainesville, FL 32611, USA.,Institute for Cell &Tissue Science and Engineering, University of Florida, Gainesville, FL 32611, USA.,National Cancer Institute-Physical Science Oncology Center, Gainesville, FL 32611, USA
| |
Collapse
|
30
|
Sweat RS, Azimi MS, Suarez-Martinez AD, Katakam P, Murfee WL. Lysophosphatidic acid does not cause blood/lymphatic vessel plasticity in the rat mesentery culture model. Physiol Rep 2016; 4:4/13/e12857. [PMID: 27401461 PMCID: PMC4945839 DOI: 10.14814/phy2.12857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/16/2016] [Indexed: 12/27/2022] Open
Abstract
Understanding the mechanisms behind endothelial cell identity is crucial for the goal of manipulating microvascular networks. Lysophosphatidic acid (LPA) and serum stimulation have been suggested to induce a lymphatic identity in blood endothelial cells in vitro. The objective of this study was to determine if LPA or serum induces blood‐to‐lymphatic vessel phenotypic transition in microvascular networks. The rat mesentery culture model was used to observe the effect of stimulation on blood and lymphatic microvascular networks ex vivo. Vascularized mesenteric tissues were harvested from adult Wistar rats and cultured with LPA or 10% serum for up to 5 days. Tissues were then immunolabeled with PECAM to identify blood vessels and LYVE‐1 or Prox1 to identify lymphatic vessels. We show that while LPA caused capillary sprouting and increased vascular length density in adult microvascular networks, LPA did not cause a blood‐to‐lymphatic phenotypic transition. The results suggest that LPA is not sufficient to cause blood endothelial cells to adopt a lymphatic identity in adult microvascular networks. Similarly, serum stimulation caused robust angiogenesis and increased lymphatic/blood vessel connections, yet did not induce a blood‐to‐lymphatic phenotypic transition. Our study highlights an understudied area of lymphatic research and warrants future investigation into the mechanisms responsible for the maintenance of blood and lymphatic vessel identity.
Collapse
Affiliation(s)
- Richard S Sweat
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Mohammad S Azimi
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | | | - Prasad Katakam
- Department of Pharmacology, Tulane University, New Orleans, Louisiana
| | - Walter L Murfee
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| |
Collapse
|
31
|
Pham PH, Vo NTK, Tan EJH, Russell S, Jones G, Lumsden JS, Bols NC. Development of an Atlantic salmon heart endothelial cell line (ASHe) that responds to lysophosphatidic acid (LPA). In Vitro Cell Dev Biol Anim 2016; 53:20-32. [PMID: 27586265 DOI: 10.1007/s11626-016-0077-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 07/22/2016] [Indexed: 12/16/2022]
Abstract
As diseases and abnormalities of the heart can interfere with the aquaculture of Atlantic salmon, the heart was investigated as a source of cell lines that could be used to study the cellular basis of these conditions. An Atlantic salmon heart endothelial cell line, ASHe, was developed and characterized for growth properties, endothelial cell characteristics, and responsiveness to lysophosphatidic acid (LPA). AHSe cells stained negative for senescence associated ß-galactosidase and grew well in 10 and 20% FBS/L15 at high cell density, but not in L15 medium supplemented with calf serum. It displayed many endothelial cell-like characteristics including a cobblestone morphology, capillary-like structures formation on Matrigel, and expression of von Willebrand factor and endothelial cell-related tight junction proteins ZO-1, claudin 3, and claudin 5. ASHe cells responded to the cardiovascular modulator, LPA, in two contrasting ways. LPA at 5 and 25 μM inhibited the ability of ASHe cells to heal a wound but stimulated their proliferation, especially as evaluated by colony formation in low-density cultures. The enhancement of proliferation by LPA parallels what has been observed previously in mammalian endothelial cell cultures exposed to LPA, whereas the LPA slowing of ASHe cell migration contrasted with the LPA-enhanced migration of some mammalian cells. Therefore, this cell line is a potentially useful model for future comparative studies on piscine and mammalian cardiovascular cell biology and for studies on diseases of Atlantic salmon in aquaculture.
Collapse
Affiliation(s)
- Phuc H Pham
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Nguyen T K Vo
- Department of Medical Physics and Applied Radiation Sciences, McMaster University, Hamilton, ON, Canada
| | | | - Spencer Russell
- Department of Fisheries and Aquaculture, Vancouver Island University, Nanaimo, BC, Canada
| | | | - John S Lumsden
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Niels C Bols
- Department of Biology, University of Waterloo, Waterloo, ON, Canada.
| |
Collapse
|
32
|
Chhokar V, Tucker AL. Angiogenesis: Basic Mechanisms and Clinical Applications. Semin Cardiothorac Vasc Anesth 2016. [DOI: 10.1177/108925320300700304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The development and maintenance of an adequate vascular supply is critical for the viability of normal and neoplastic tissues. Angiogenesis, the development of new blood vessels from preexisting capillary networks, plays an important role in a number of physiologic and pathologic processes, including reproduction, wound repair, inflammatory diseases, and tumor growth. Angiogenesis involves sequential steps that are triggered in response to angiogenic growth factors released by inflammatory, mesenchymal, or tumor cells that act as ligands for endothelial cell receptor tyrosine kinases. Stimulated endothelial cells detach from neighboring cells and migrate, proliferate, and form tubes. The immature tubes are subsequently invested and stabilized by pericytes or smooth muscle cells. Angiogenesis depends upon complex interactions among various classes of molecules, including adhesion molecules, proteases, structural proteins, cell surface receptors, and growth factors. The therapeutic manipulation of angiogenesis targeted against ischemic and neoplastic diseases has been investigated in preclinical animal models and in clinical trials. Proangiogenic trials that have stimulated vessel growth in ischemic coronary or peripheral tissues through expression, delivery, or stimulated release of growth factors have shown efficacy in animal models and mixed results in human clinical trials. Antiangiogenic trials have used strategies to block the function of molecules critical for new vessel growth or maturation in the treatment of a variety of malignancies, mostly with results less encouraging than those seen in preclinical models. Pro-and antiangiogenic clinical trials demonstrate that strategies for optimal drug delivery, dosing schedules, patient selection, and endpoint measurements need further investigation and refinement before the therapeutic manipulation of angiogenesis will realize its full clinical potential.
Collapse
Affiliation(s)
- Vikram Chhokar
- Department of Internal Medicine, Salem VA Health System, Roanoke, Virginia
| | - Amy L. Tucker
- Department of Internal Medicine, Cardiovascular Division; Cardiovascular Research Center; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
33
|
Han JK, Kim BK, Won JY, Shin Y, Choi SB, Hwang I, Kang J, Lee HJ, Koh SJ, Lee J, Hur J, Cho HJ, Chae IH, Oh BH, Park YB, Kim HS. Interaction between platelets and endothelial progenitor cells via LPA-Edg-2 axis is augmented by PPAR-δ activation. J Mol Cell Cardiol 2016; 97:266-77. [DOI: 10.1016/j.yjmcc.2016.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 05/05/2016] [Accepted: 06/01/2016] [Indexed: 11/26/2022]
|
34
|
Thorlakson HH, Schreurs O, Schenck K, Blix IJS. Lysophosphatidic acid regulates adhesion molecules and enhances migration of human oral keratinocytes. Eur J Oral Sci 2016; 124:164-71. [PMID: 26913569 DOI: 10.1111/eos.12255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2016] [Indexed: 12/20/2022]
Abstract
Oral keratinocytes are connected via cell-to-cell adhesions to protect underlying tissues from physical and bacterial damage. Lysophosphatidic acids (LPAs) are a family of phospholipid mediators that have the ability to regulate gene expression, cytoskeletal rearrangement, and cytokine/chemokine secretion, which mediate proliferation, migration, and differentiation. Several forms of LPA are found in saliva and gingival crevicular fluid, but it is unknown how they affect human oral keratinocytes (HOK). The aim of the present study was therefore to examine how different LPA forms affect the expression of adhesion molecules and the migration and proliferation of HOK. Keratinocytes were isolated from gingival biopsies obtained from healthy donors and challenged with different forms of LPA. Quantitative real-time RT-PCR, immunocytochemistry, and flow cytometry were used to analyze the expression of adhesion molecules. Migration and proliferation assays were performed. Lysophosphatidic acids strongly promoted expression of E-cadherin and occludin mRNAs and translocation of E-cadherin protein from the cytoplasm to the membrane. Occludin and claudin-1 proteins were up-regulated by LPA. Migration of HOK in culture was increased, but proliferation was reduced, by the addition of LPA. This indicates that LPA can have a role in the regulation of the oral epithelial barrier by increasing the expression of adhesion molecules of HOK, by promotion of migration and by inhibition of proliferation.
Collapse
Affiliation(s)
- Hong H Thorlakson
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.,Department of Periodontology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Olav Schreurs
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Karl Schenck
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Inger J S Blix
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.,Department of Periodontology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
35
|
bFGF-Regulating MAPKs Are Involved in High Glucose-Mediated ROS Production and Delay of Vascular Endothelial Cell Migration. PLoS One 2015; 10:e0144495. [PMID: 26642060 PMCID: PMC4671674 DOI: 10.1371/journal.pone.0144495] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/19/2015] [Indexed: 01/09/2023] Open
Abstract
High blood sugar is a symptom of diabetes mellitus (DM). Vascular endothelial cells (VECs) directly contact the blood and are damaged when blood sugar levels are high. However, the molecular mechanism underlying this process remains elusive. To analyze the effects of DM on migration, we simulated DM by applying high glucose (HG) to the human VEC. HG delayed cell migration and induced phosphorylation of MAPKs (JNK and ERK). By contrast, in presence of bFGF, cell migration was promoted and MAPK phosphorylation levels were reduced. Furthermore, treatment with JNK and ERK inhibitors rescued HG-mediated delay of cell migration. Molecular and cell biological studies demonstrated that HG increased ROS production, whereas treatment with bFGF or JNK/ERK inhibitors blocked HG-induced ROS accumulation. Addition of MnTMPyP, a ROS scavenger, reduced HG-induced ROS production and accelerated cell migration, suggesting that the influence of HG on bFGF-MAPK signaling causes accumulation of ROS, which in turn regulate cell migration. This is the first study to elucidate the molecular mechanism of HG-mediated VEC migration; these findings could facilitate the development of novel therapies for DM.
Collapse
|
36
|
Mahajan-Thakur S, Böhm A, Jedlitschky G, Schrör K, Rauch BH. Sphingosine-1-Phosphate and Its Receptors: A Mutual Link between Blood Coagulation and Inflammation. Mediators Inflamm 2015; 2015:831059. [PMID: 26604433 PMCID: PMC4641948 DOI: 10.1155/2015/831059] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/26/2015] [Accepted: 09/30/2015] [Indexed: 02/02/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a versatile lipid signaling molecule and key regulator in vascular inflammation. S1P is secreted by platelets, monocytes, and vascular endothelial and smooth muscle cells. It binds specifically to a family of G-protein-coupled receptors, S1P receptors 1 to 5, resulting in downstream signaling and numerous cellular effects. S1P modulates cell proliferation and migration, and mediates proinflammatory responses and apoptosis. In the vascular barrier, S1P regulates permeability and endothelial reactions and recruitment of monocytes and may modulate atherosclerosis. Only recently has S1P emerged as a critical mediator which directly links the coagulation factor system to vascular inflammation. The multifunctional proteases thrombin and FXa regulate local S1P availability and interact with S1P signaling at multiple levels in various vascular cell types. Differential expression patterns and intracellular signaling pathways of each receptor enable S1P to exert its widespread functions. Although a vast amount of information is available about the functions of S1P and its receptors in the regulation of physiological and pathophysiological conditions, S1P-mediated mechanisms in the vasculature remain to be elucidated. This review summarizes recent findings regarding the role of S1P and its receptors in vascular wall and blood cells, which link the coagulation system to inflammatory responses in the vasculature.
Collapse
Affiliation(s)
- Shailaja Mahajan-Thakur
- Institut für Pharmakologie, Universitätsmedizin Greifswald, Felix-Hausdorf Strasse 3, 17487 Greifswald, Germany
| | - Andreas Böhm
- Institut für Pharmakologie, Universitätsmedizin Greifswald, Felix-Hausdorf Strasse 3, 17487 Greifswald, Germany
| | - Gabriele Jedlitschky
- Institut für Pharmakologie, Universitätsmedizin Greifswald, Felix-Hausdorf Strasse 3, 17487 Greifswald, Germany
| | - Karsten Schrör
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Bernhard H. Rauch
- Institut für Pharmakologie, Universitätsmedizin Greifswald, Felix-Hausdorf Strasse 3, 17487 Greifswald, Germany
| |
Collapse
|
37
|
Hattori Y, Kim H, Tsuboi N, Yamamoto A, Akiyama S, Shi Y, Katsuno T, Kosugi T, Ueda M, Matsuo S, Maruyama S. Therapeutic Potential of Stem Cells from Human Exfoliated Deciduous Teeth in Models of Acute Kidney Injury. PLoS One 2015; 10:e0140121. [PMID: 26509261 PMCID: PMC4625005 DOI: 10.1371/journal.pone.0140121] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 09/21/2015] [Indexed: 12/15/2022] Open
Abstract
Background Acute kidney injury (AKI) is a critical condition associated with high mortality. However, the available treatments for AKI are limited. Stem cells from human exfoliated deciduous teeth (SHED) have recently gained attention as a novel source of stem cells. The purpose of this study was to clarify whether SHED have a therapeutic effect on AKI induced by ischemia-reperfusion injury. Methods The left renal artery and vein of the mice were clamped for 20 min to induce ischemia. SHED, bone marrow derived mesenchymal stem cells (BMMSC) or phosphate-buffered saline (control) were administered into the subrenal capsule. To confirm the potency of SHED in vitro, H2O2 stimulation assays and scratch assays were performed. Results The serum creatinine and blood urea nitrogen levels of the SHED group were significantly lower than those of the control group, while BMMSC showed no therapeutic effect. Infiltration of macrophages and neutrophils in the kidney was significantly attenuated in mice treated with SHED. Cytokine levels (MIP-2, IL-1β, and MCP-1) in mice kidneys were significantly reduced in the SHED group. In in vitro experiments, SHED significantly decreased MCP-1 secretion in tubular epithelial cells (TEC) stimulated with H2O2. In addition, SHED promoted wound healing in the scratch assays, which was blunted by anti-HGF antibodies. Discussion SHED attenuated the levels of inflammatory cytokines and improved kidney function in AKI induced by IRI. SHED secreted factors reduced MCP-1 and increased HGF expression, which promoted wound healing. These results suggest that SHED might provide a novel stem cell resource, which can be applied for the treatment of ischemic kidney injury.
Collapse
Affiliation(s)
- Yuka Hattori
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Hangsoo Kim
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Naotake Tsuboi
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Akihito Yamamoto
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Shinichi Akiyama
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Yiqin Shi
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Takayuki Katsuno
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Tomoki Kosugi
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Minoru Ueda
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Seiichi Matsuo
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Internal Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
- * E-mail:
| |
Collapse
|
38
|
Hwang SH, Lee BH, Choi SH, Kim HJ, Won KJ, Lee HM, Rhim H, Kim HC, Nah SY. Effects of gintonin on the proliferation, migration, and tube formation of human umbilical-vein endothelial cells: involvement of lysophosphatidic-acid receptors and vascular-endothelial-growth-factor signaling. J Ginseng Res 2015; 40:325-333. [PMID: 27746684 PMCID: PMC5052429 DOI: 10.1016/j.jgr.2015.10.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/08/2015] [Accepted: 10/19/2015] [Indexed: 11/22/2022] Open
Abstract
Background Ginseng extracts are known to have angiogenic effects. However, to date, only limited information is available on the molecular mechanism underlying the angiogenic effects and the main components of ginseng that exert these effects. Human umbilical-vein endothelial cells (HUVECs) are used as an in vitro model for screening therapeutic agents that promote angiogenesis and wound healing. We recently isolated gintonin, a novel ginseng-derived lysophosphatidic acid (LPA) receptor ligand, from ginseng. LPA plays a key role in angiogenesis and wound healing. Methods In the present study, we investigated the in vitro effects of gintonin on proliferation, migration, and tube formation of HUVECs, which express endogenous LPA1/3 receptors. Results Gintonin stimulated proliferation and migration of HUVECs. The LPA1/3 receptor antagonist, Ki16425, short interfering RNA against LPA1 or LPA3 receptor, and the Rho kinase inhibitor, Y-27632, significantly decreased the gintonin-induced proliferation, migration, and tube formation of HUVECs, which indicates the involvement of LPA receptors and Rho kinase activation. Further, gintonin increased the release of vascular endothelial growth factors from HUVECs. The cyclooxygenase-2 inhibitor NS-398, nuclear factor kappa B inhibitor BAY11-7085, and c-Jun N-terminal kinase inhibitor SP600125 blocked the gintonin-induced migration, which shows the involvement of cyclooxygenase-2, nuclear factor kappa B, and c-Jun N-terminal kinase signaling. Conclusion The gintonin-mediated proliferation, migration, and vascular-endothelial-growth-factor release in HUVECs via LPA-receptor activation may be one of in vitro mechanisms underlying ginseng-induced angiogenic and wound-healing effects.
Collapse
Affiliation(s)
- Sung-Hee Hwang
- Department of Pharmaceutical Engineering, College of Health Sciences, Sangji University, Wonju, Korea
| | - Byung-Hwan Lee
- Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, Korea
| | - Sun-Hye Choi
- Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, Korea
| | - Hyeon-Joong Kim
- Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, Korea
| | - Kyung Jong Won
- Department of Physiology, School of Medicine, Konkuk University, Chungju, Korea
| | - Hwan Myung Lee
- Department of Cosmetic Science, College of Natural Science, Hoseo University, Asan, Korea
| | - Hyewon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul, Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Korea
| | - Seung-Yeol Nah
- Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, Korea
| |
Collapse
|
39
|
Durham JT, Dulmovits BM, Cronk SM, Sheets AR, Herman IM. Pericyte chemomechanics and the angiogenic switch: insights into the pathogenesis of proliferative diabetic retinopathy? Invest Ophthalmol Vis Sci 2015; 56:3441-59. [PMID: 26030100 DOI: 10.1167/iovs.14-13945] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
PURPOSE To establish the regulatory roles that pericytes have in coordinating retinal endothelial cell (EC) growth and angiogenic potential. METHODS Pericytes were derived from donor diabetic (DHuRP) or normal (NHuRP) human retinae, and characterized using vascular markers, coculture, contraction, morphogenesis, and proliferation assays. To investigate capillary "cross-talk," pericyte-endothelial coculture growth, and connexin-43 (Cx43) expression assays were performed. Paracrine effects were examined via treating EC with pericyte-derived conditioned media (CM) in proliferation, angiogenesis, and angiocrine assays. The effects of sphingosine 1-phosphate (S1P) were assessed using receptor antagonists. RESULTS The DHuRP exhibit unique proliferative and morphologic properties, reflecting distinctive cytoskeletal and isoactin expression patterns. Unlike NHuRP, DHuRP are unable to sustain EC growth arrest in coculture and display reduced Cx43 expression. Further, CM from DHuRP (DPCM) markedly stimulates EC proliferation and tube formation. Treatment with S1P receptor antagonists mitigates DPCM growth-promotion in EC and S1P-mediated pericyte contraction. Angiocrine assays on normal and diabetic pericyte secretomes reveal factors involved in angiogenic control, inflammation, and metabolism. CONCLUSIONS Effects from the diabetic microenvironment appear sustainable in cell culture: pericytes derived from diabetic donor eyes seemingly possess a "metabolic memory" in vitro, which may be linked to original donor health status. Diabetes- and pericyte-dependent effects on EC growth and angiogenesis may reflect alterations in bioactive lipid, angiocrine, and chemomechanical signaling. Altogether, our results suggest that diabetes alters pericyte contractile phenotype and cytoskeletal signaling, which ultimately may serve as a key, initiating event required for retinal endothelial reproliferation, angiogenic activation, and the pathological neovascularization accompanying proliferative diabetic retinopathy.
Collapse
|
40
|
KANG SANGJIN, HAN JUHEE, SONG SEUNGYONG, KIM WONSERK, SHIN SOYOUNG, KIM JIHYE, AHN HYOSUN, JEONG JINHYUN, HWANG SUNGJOO, SUNG JONGHYUK. Lysophosphatidic acid increases the proliferation and migration of adipose-derived stem cells via the generation of reactive oxygen species. Mol Med Rep 2015; 12:5203-10. [DOI: 10.3892/mmr.2015.4023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 06/15/2015] [Indexed: 11/06/2022] Open
|
41
|
Kawashima Y, Kushida N, Kokubun S, Ogawa S, Shiomi H, Ishibashi K, Aikawa K, Ikegami K, Nomiya M, Yamaguchi O. Possible effect of lysophosphatidic acid on cell proliferation and involvement of lysophosphatidic acid and lysophosphatidic acid receptors in mechanical stretch-induced mitogen-activated protein kinase. Int J Urol 2015; 22:778-84. [DOI: 10.1111/iju.12799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 03/26/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Yohei Kawashima
- Department of Urology; Fukushima Medical University School of Medicine; Fukushima Japan
| | - Nobuhiro Kushida
- Department of Urology; Fukushima Medical University School of Medicine; Fukushima Japan
| | - Shuko Kokubun
- Department of Urology; Fukushima Medical University School of Medicine; Fukushima Japan
| | - Soichiro Ogawa
- Department of Urology; Fukushima Medical University School of Medicine; Fukushima Japan
| | - Homare Shiomi
- Department of Urology; Fukushima Medical University School of Medicine; Fukushima Japan
| | - Kei Ishibashi
- Department of Urology; Fukushima Medical University School of Medicine; Fukushima Japan
| | - Ken Aikawa
- Department of Urology; Fukushima Medical University School of Medicine; Fukushima Japan
| | - Kentaro Ikegami
- Division of Bioengineering and LUTD Research; Nihon University College of Engineering; Koriyama Japan
| | - Masanori Nomiya
- Division of Bioengineering and LUTD Research; Nihon University College of Engineering; Koriyama Japan
| | - Osamu Yamaguchi
- Division of Bioengineering and LUTD Research; Nihon University College of Engineering; Koriyama Japan
| |
Collapse
|
42
|
González de San Román E, Manuel I, Giralt MT, Chun J, Estivill-Torrús G, Rodríguez de Fonseca F, Santín LJ, Ferrer I, Rodríguez-Puertas R. Anatomical location of LPA1 activation and LPA phospholipid precursors in rodent and human brain. J Neurochem 2015; 134:471-85. [PMID: 25857358 DOI: 10.1111/jnc.13112] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/02/2015] [Accepted: 03/31/2015] [Indexed: 12/29/2022]
Abstract
Lysophosphatidic acid (LPA) is a signaling molecule that binds to six known G protein-coupled receptors: LPA1 -LPA6 . LPA evokes several responses in the CNS, including cortical development and folding, growth of the axonal cone and its retraction process. Those cell processes involve survival, migration, adhesion proliferation, differentiation, and myelination. The anatomical localization of LPA1 is incompletely understood, particularly with regard to LPA binding. Therefore, we have used functional [(35) S]GTPγS autoradiography to verify the anatomical distribution of LPA1 binding sites in adult rodent and human brain. The greatest activity was observed in myelinated areas of the white matter such as corpus callosum, internal capsule and cerebellum. MaLPA1 -null mice (a variant of LPA1 -null) lack [(35) S]GTPγS basal binding in white matter areas, where the LPA1 receptor is expressed at high levels, suggesting a relevant role of the activity of this receptor in the most myelinated brain areas. In addition, phospholipid precursors of LPA were localized by MALDI-IMS in both rodent and human brain slices identifying numerous species of phosphatides and phosphatidylcholines. Both phosphatides and phosphatidylcholines species represent potential LPA precursors. The anatomical distribution of these precursors in rodent and human brain may indicate a metabolic relationship between LPA and LPA1 receptors. Lysophosphatidic acid (LPA) is a signaling molecule that binds to six known G protein-coupled receptors (GPCR), LPA1 to LPA6 . LPA evokes several responses in the central nervous system (CNS), including cortical development and folding, growth of the axonal cone and its retraction process. We used functional [(35) S]GTPγS autoradiography to verify the anatomical distribution of LPA1 -binding sites in adult rodent and human brain. The distribution of LPA1 receptors in rat, mouse and human brains show the highest activity in white matter myelinated areas. The basal and LPA-evoked activities are abolished in MaLPA1 -null mice. The phospholipid precursors of LPA are localized by MALDI-IMS. The anatomical distribution of LPA precursors in rodent and human brain suggests a relationship with functional LPA1 receptors.
Collapse
Affiliation(s)
| | - Iván Manuel
- Department of Pharmacology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - María Teresa Giralt
- Department of Pharmacology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Jerold Chun
- Molecular and Cellular Neuroscience Department, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California, USA
| | - Guillermo Estivill-Torrús
- UGC Intercentros de Neurociencias y UGC de Salud Mental, Instituto de Investigación Biomédica de Malaga (IBIMA), Hospitales Universitarios Regional de Málaga y Virgen de la Victoria, Universidad de Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- UGC Intercentros de Neurociencias y UGC de Salud Mental, Instituto de Investigación Biomédica de Malaga (IBIMA), Hospitales Universitarios Regional de Málaga y Virgen de la Victoria, Universidad de Málaga, Spain
| | - Luis Javier Santín
- Departmento de Psicobiología y Metodología de las Ciencias del Comportamiento. Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad of Málaga, Málaga, Spain
| | - Isidro Ferrer
- Institute of Neuropathology, University Hospital Bellvitge, University of Barcelona, Ciberned, Spain
| | - Rafael Rodríguez-Puertas
- Department of Pharmacology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Leioa, Spain
| |
Collapse
|
43
|
Williams PA, Stilhano RS, To VP, Tran L, Wong K, Silva EA. Hypoxia augments outgrowth endothelial cell (OEC) sprouting and directed migration in response to sphingosine-1-phosphate (S1P). PLoS One 2015; 10:e0123437. [PMID: 25875493 PMCID: PMC4398361 DOI: 10.1371/journal.pone.0123437] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 02/20/2015] [Indexed: 12/26/2022] Open
Abstract
Therapeutic angiogenesis provides a promising approach to treat ischemic cardiovascular diseases through the delivery of proangiogenic cells and/or molecules. Outgrowth endothelial cells (OECs) are vascular progenitor cells that are especially suited for therapeutic strategies given their ease of noninvasive isolation from umbilical cord or adult peripheral blood and their potent ability to enhance tissue neovascularization. These cells are recruited to sites of vascular injury or tissue ischemia and directly incorporate within native vascular endothelium to participate in neovessel formation. A better understanding of how OEC activity may be boosted under hypoxia with external stimulation by proangiogenic molecules remains a challenge to improving their therapeutic potential. While vascular endothelial growth factor (VEGF) is widely established as a critical factor for initiating angiogenesis, sphingosine-1-phosphate (S1P), a bioactive lysophospholipid, has recently gained great enthusiasm as a potential mediator in neovascularization strategies. This study tests the hypothesis that hypoxia and the presence of VEGF impact the angiogenic response of OECs to S1P stimulation in vitro. We found that hypoxia altered the dynamically regulated S1P receptor 1 (S1PR1) expression on OECs in the presence of S1P (1.0 μM) and/or VEGF (1.3 nM). The combined stimuli of S1P and VEGF together promoted OEC angiogenic activity as assessed by proliferation, wound healing, 3D sprouting, and directed migration under both normoxia and hypoxia. Hypoxia substantially augmented the response to S1P alone, resulting in ~6.5-fold and ~25-fold increases in sprouting and directed migration, respectively. Overall, this report highlights the importance of establishing hypoxic conditions in vitro when studying ischemia-related angiogenic strategies employing vascular progenitor cells.
Collapse
Affiliation(s)
- Priscilla A. Williams
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Roberta S. Stilhano
- Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | - Vivian P. To
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Lyndon Tran
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, California, United States of America
| | - Kevin Wong
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Eduardo A. Silva
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
44
|
Hsiao CM, Wu YS, Nan FH, Huang SL, Chen L, Chen SN. Immunomodulator 'mushroom beta glucan' induces Wnt/β catenin signalling and improves wound recovery in tilapia and rat skin: a histopathological study. Int Wound J 2015; 13:1116-1128. [PMID: 25857741 DOI: 10.1111/iwj.12427] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/03/2015] [Accepted: 01/24/2015] [Indexed: 12/18/2022] Open
Abstract
The present study aims to investigate the effects of mushroom beta glucan (MBG) on wound recovery in partial hepatectomy (PH) in Nile tilapia (Oreochromis niloticus) and in rat skin wound healing examination. Following PH, we focussed on the effects on liver repair ability using in vitro and in vivo tests. In vitro, we examined whether the MBG has an impact on liver cell proliferation, mainly through 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT) assays and bromodeoxyuridine (BrdU) cell proliferation assay detection method. Results showed that MBG treatment was remarkable in enhancing cell proliferation of hepatocytes and in maintaining the cellular viability. Immunohistochemical staining to analyse Wnt/β-catenin signalling also showed that MBG has the effect of promoting cell proliferation of liver tissues after PH surgery.
Collapse
Affiliation(s)
- Chien-Mei Hsiao
- Institute of Fisheries Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Sheng Wu
- Institute of Fisheries Science, National Taiwan University, Taipei, Taiwan
| | - Fan-Hua Nan
- Department of Aquaculture, National Taiwan Ocean University, Keelung, Taiwan
| | - Shih-Ling Huang
- Freshwater Aquaculture Research Center, Fisheries Research Institute, Council of Agriculture, Changhua, Taiwan
| | - Lynette Chen
- Oregon Health and Science University Hospital Oregon, Oregon, OR, USA
| | - Shiu-Nan Chen
- Institute of Fisheries Science, National Taiwan University, Taipei, Taiwan.,College of Life Science, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
45
|
Chou CH, Lai SL, Ho CM, Lin WH, Chen CN, Lee PH, Peng FC, Kuo SH, Wu SY, Lai HS. Lysophosphatidic acid alters the expression profiles of angiogenic factors, cytokines, and chemokines in mouse liver sinusoidal endothelial cells. PLoS One 2015; 10:e0122060. [PMID: 25822713 PMCID: PMC4379007 DOI: 10.1371/journal.pone.0122060] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 02/08/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND AND AIMS Lysophosphatidic acid (LPA) is a multi-function glycerophospholipid. LPA affects the proliferation of hepatocytes and stellate cells in vitro, and in a partial hepatectomy induced liver regeneration model, the circulating LPA levels and LPA receptor (LPAR) expression levels in liver tissue are significantly changed. Liver sinusoidal endothelial cells (Lsecs) play an important role during liver regeneration. However, the effects of LPA on Lsecs are not well known. Thus, we investigated the effects of LPA on the expression profiles of angiogenic factors, cytokines, and chemokines in Lsecs. METHODS Mouse Lsecs were isolated using CD31-coated magnetic beads. The mRNA expression levels of LPAR's and other target genes were determined by quantitative RT-PCR. The protein levels of angiogenesis factors, cytokines, and chemokines were determined using protein arrays and enzyme immunoassay (EIA). Critical LPAR related signal transduction was verified by using an appropriate chemical inhibitor. RESULTS LPAR1 and LPAR3 mRNA's were expressed in mouse LPA-treated Lsecs. Treating Lsecs with a physiological level of LPA significantly enhanced the protein levels of angiogenesis related proteins (cyr61 and TIMP-1), cytokines (C5/C5a, M-CSF, and SDF-1), and chemokines (MCP-5, gp130, CCL28, and CXCL16). The LPAR1 and LPAR3 antagonist ki16425 significantly inhibited the LPA-enhanced expression of cyr61, TIMP-1, SDF-1, MCP-5, gp130, CCL28, and CXCL16, but not that of C5/C5a or M-CSF. LPA-induced C5/C5a and M-CSF expression may have been through an indirect regulation mechanism. CONCLUSION LPA regulated the expression profiles of angiogenic factors, cytokines, and chemokines in Lsecs that was mediated via LPAR1 and LPAR3 signaling. Most of the factors that were enhanced by LPA have been found to play critical roles during liver regeneration. Thus, these results may prove useful for manipulating LPA effects on liver regeneration.
Collapse
Affiliation(s)
- Chia-Hung Chou
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shou-Lun Lai
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Cheng-Maw Ho
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wen-Hsi Lin
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chiung-Nien Chen
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Po-Huang Lee
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Fu-Chuo Peng
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sung-Hsin Kuo
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Szu-Yuan Wu
- Department of Radiation-Oncology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hong-Shiee Lai
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
46
|
Stoddard NC, Chun J. Promising pharmacological directions in the world of lysophosphatidic Acid signaling. Biomol Ther (Seoul) 2015; 23:1-11. [PMID: 25593637 PMCID: PMC4286743 DOI: 10.4062/biomolther.2014.109] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/02/2014] [Accepted: 12/02/2014] [Indexed: 12/18/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a signaling lipid that binds to six known lysophosphatidic acid receptors (LPARs), named LPA1-LPA6. These receptors initiate signaling cascades relevant to development, maintenance, and healing processes throughout the body. The diversity and specificity of LPA signaling, especially in relation to cancer and autoimmune disorders, makes LPA receptor modulation an attractive target for drug development. Several LPAR-specific analogues and small molecules have been synthesized and are efficacious in attenuating pathology in disease models. To date, at least three compounds have passed phase I and phase II clinical trials for idiopathic pulmonary fibrosis and systemic sclerosis. This review focuses on the promising therapeutic directions emerging in LPA signaling toward ameliorating several diseases, including cancer, fibrosis, arthritis, hydrocephalus, and traumatic injury.
Collapse
Affiliation(s)
- Nicole C Stoddard
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037 ; Biomedical Sciences Graduate Program, University of California, San Diego, School of Medicine, La Jolla, CA 92037, USA
| | - Jerold Chun
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
47
|
Ogle ME, Sefcik LS, Awojoodu AO, Chiappa NF, Lynch K, Peirce-Cottler S, Botchwey EA. Engineering in vivo gradients of sphingosine-1-phosphate receptor ligands for localized microvascular remodeling and inflammatory cell positioning. Acta Biomater 2014; 10:4704-4714. [PMID: 25128750 PMCID: PMC4529737 DOI: 10.1016/j.actbio.2014.08.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 07/30/2014] [Accepted: 08/06/2014] [Indexed: 12/29/2022]
Abstract
Biomaterial-mediated controlled release of soluble signaling molecules is a tissue engineering approach to spatially control processes of inflammation, microvascular remodeling and host cell recruitment, and to generate biochemical gradients in vivo. Lipid mediators, such as sphingosine 1-phosphate (S1P), are recognized for their essential roles in spatial guidance, signaling and highly regulated endogenous gradients. S1P and pharmacological analogs such as FTY720 are therapeutically attractive targets for their critical roles in the trafficking of cells between blood and tissue spaces, both physiologically and pathophysiologically. However, the interaction of locally delivered sphingolipids with the complex metabolic networks controlling the flux of lipid species in inflamed tissue has yet to be elucidated. In this study, complementary in vitro and in vivo approaches are investigated to identify relationships between polymer composition, drug release kinetics, S1P metabolic activity, signaling gradients and spatial positioning of circulating cells around poly(lactic-co-glycolic acid) biomaterials. Results demonstrate that biomaterial-based gradients of S1P are short-lived in the tissue due to degradation by S1P lyase, an enzyme that irreversibly degrades intracellular S1P. On the other hand, in vivo gradients of the more stable compound, FTY720, enhance microvascular remodeling by selectively recruiting an anti-inflammatory subset of monocytes (S1P3(high)) to the biomaterial. Results highlight the need to better understand the endogenous balance of lipid import/export machinery and lipid kinase/phosphatase activity in order to design biomaterial products that spatially control the innate immune environment to maximize regenerative potential.
Collapse
Affiliation(s)
- Molly E. Ogle
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
| | - Lauren S. Sefcik
- Department of Chemical & Biomolecular Engineering, Lafayette College, 740 High Street, Easton, PA 18042
| | - Anthony O. Awojoodu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
| | - Nathan F. Chiappa
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
| | - Kevin Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22903
| | - Shayn Peirce-Cottler
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903
| | - Edward A. Botchwey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 315 Ferst Drive, Atlanta, GA 30332
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903
| |
Collapse
|
48
|
Zhou Z, Li X, Yang B, Jiang D. Relationship between lysophosphatidic acid and matrix metalloproteinase-9 plasma concentrations and carotid atheromatous plaque stability in patients with cerebral infarction. J Int Med Res 2014; 42:669-76. [PMID: 24781723 DOI: 10.1177/0300060514526567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/10/2014] [Indexed: 11/15/2022] Open
Abstract
Objective: To investigate the correlation between plasma lysophosphatidic acid (LPA), matrix metalloproteinase (MMP)-9 and carotid atheromatous plaque stability in patients with cerebral infarction. Method: Patients with cerebral infarction underwent carotid artery duplex ultrasonography and transcranial Doppler monitoring for detection of microemboli. Patients were stratified by plaque type (no plaque, intima thickening, unstable plaque, stable plaque) and presence or absence of micoremboli. Plasma LPA and MMP-9 were quantified. Result: LPA and MMP-9 concentrations were significantly higher and microemboli positivity was significantly more common in patients with unstable plaque ( n = 21) than the other three groups (intima thickening, n = 16; stable plaque n = 41; no plaque; n = 12). There was a significant positive correlation between LPA and MMP-9 concentrations. Conclusion: Plasma LPA and MMP-9 concentrations may be useful biomarkers in the clinical identification and prediction of unstable plaque, and in guiding treatment.
Collapse
Affiliation(s)
- Zhibin Zhou
- Department of Neurology, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Xiaohao Li
- Department of Emergency Medicine, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Bo Yang
- Department of Cardiology, Zhongshan Hospital of Hubei, Wuhan, China
| | - Dan Jiang
- Department of Neurology, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
49
|
Yung YC, Stoddard NC, Chun J. LPA receptor signaling: pharmacology, physiology, and pathophysiology. J Lipid Res 2014; 55:1192-214. [PMID: 24643338 DOI: 10.1194/jlr.r046458] [Citation(s) in RCA: 530] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Indexed: 12/18/2022] Open
Abstract
Lysophosphatidic acid (LPA) is a small ubiquitous lipid found in vertebrate and nonvertebrate organisms that mediates diverse biological actions and demonstrates medicinal relevance. LPA's functional roles are driven by extracellular signaling through at least six 7-transmembrane G protein-coupled receptors. These receptors are named LPA1-6 and signal through numerous effector pathways activated by heterotrimeric G proteins, including Gi/o, G12/13, Gq, and Gs LPA receptor-mediated effects have been described in numerous cell types and model systems, both in vitro and in vivo, through gain- and loss-of-function studies. These studies have revealed physiological and pathophysiological influences on virtually every organ system and developmental stage of an organism. These include the nervous, cardiovascular, reproductive, and pulmonary systems. Disturbances in normal LPA signaling may contribute to a range of diseases, including neurodevelopmental and neuropsychiatric disorders, pain, cardiovascular disease, bone disorders, fibrosis, cancer, infertility, and obesity. These studies underscore the potential of LPA receptor subtypes and related signaling mechanisms to provide novel therapeutic targets.
Collapse
Affiliation(s)
- Yun C Yung
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| | - Nicole C Stoddard
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037 Biomedical Sciences Graduate Program, University of California, San Diego School of Medicine, La Jolla, CA 92037
| | - Jerold Chun
- Department of Molecular and Cellular Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
50
|
Kim H, Mizuno M, Furuhashi K, Katsuno T, Ozaki T, Yasuda K, Tsuboi N, Sato W, Suzuki Y, Matsuo S, Ito Y, Maruyama S. Rat adipose tissue-derived stem cells attenuate peritoneal injuries in rat zymosan-induced peritonitis accompanied by complement activation. Cytotherapy 2013; 16:357-68. [PMID: 24364907 DOI: 10.1016/j.jcyt.2013.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 09/02/2013] [Accepted: 10/22/2013] [Indexed: 01/22/2023]
Abstract
BACKGROUND AIMS In patients receiving peritoneal dialysis, fungal or yeast peritonitis has a poor prognosis. In rat peritoneum with mechanical scraping, severe peritonitis can be induced by zymosan, a component of yeast (Zy/scraping peritonitis). Administration of rat adipose tissue-derived stromal cells (ASCs) potentially can improve several tissue injuries. The present study investigated whether rat ASCs could improve peritoneal inflammation in Zy/scraping peritonitis. METHODS Rat ASCs were injected intraperitoneally on a daily basis in rats with Zy/scraping peritonitis. RESULTS Peritoneal inflammation accompanied by accumulation of inflammatory cells and complement deposition was suppressed by day 5 after injection of rat ASCs. The peritoneal mesothelial layer in Zy/scraping peritonitis with rat ASC treatment was restored compared with the peritoneal mesothelial layer without rat ASC treatment. Injected rat ASCs co-existed with mesothelial cells in the sub-peritoneal layer. In vitro assays showed increased cellular proliferation of rat mesothelial cells combined with rat ASCs by co-culture assays, confirming that fluid factors from rat ASCs might play some role in facilitating the recovery of rat mesothelial cells. Hepatocyte growth factor was released from rat ASCs, and administration of recombinant hepatocyte growth factor increased rat mesothelial cell proliferation. CONCLUSIONS Because the peritoneal mesothelium shows strong expression of membrane complement regulators such as Crry, CD55 and CD59, restoration of the mesothelial cell layer by rat ASCs might prevent deposition of complement activation products and ameliorate peritoneal injuries. This study suggests the therapeutic possibilities of intraperitoneal rat ASC injection to suppress peritoneal inflammation by restoring the mesothelial layer and decreasing complement activation in fungal or yeast peritonitis.
Collapse
Affiliation(s)
- Hangsoo Kim
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masashi Mizuno
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Kazuhiro Furuhashi
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takayuki Katsuno
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takenori Ozaki
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kaoru Yasuda
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naotake Tsuboi
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Waichi Sato
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Suzuki
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seiichi Matsuo
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiko Ito
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan; Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoichi Maruyama
- Division of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|