1
|
Jiang ZJ, Gong LW. The SphK1/S1P Axis Regulates Synaptic Vesicle Endocytosis via TRPC5 Channels. J Neurosci 2023; 43:3807-3824. [PMID: 37185099 PMCID: PMC10217994 DOI: 10.1523/jneurosci.1494-22.2023] [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: 08/03/2022] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Sphingosine-1-phosphate (S1P), a bioactive sphingolipid concentrated in the brain, is essential for normal brain functions, such as learning and memory and feeding behaviors. Sphingosine kinase 1 (SphK1), the primary kinase responsible for S1P production in the brain, is abundant within presynaptic terminals, indicating a potential role of the SphK1/S1P axis in presynaptic physiology. Altered S1P levels have been highlighted in many neurologic diseases with endocytic malfunctions. However, it remains unknown whether the SphK1/S1P axis may regulate synaptic vesicle endocytosis in neurons. The present study evaluates potential functions of the SphK1/S1P axis in synaptic vesicle endocytosis by determining effects of a dominant negative catalytically inactive SphK1. Our data for the first time identify a critical role of the SphK1/S1P axis in endocytosis in both neuroendocrine chromaffin cells and neurons from mice of both sexes. Furthermore, our Ca2+ imaging data indicate that the SphK1/S1P axis may be important for presynaptic Ca2+ increases during prolonged stimulations by regulating the Ca2+ permeable TRPC5 channels, which per se regulate synaptic vesicle endocytosis. Collectively, our data point out a critical role of the regulation of TRPC5 by the SphK1/S1P axis in synaptic vesicle endocytosis.SIGNIFICANCE STATEMENT Sphingosine kinase 1 (SphK1), the primary kinase responsible for brain sphingosine-1-phosphate (S1P) production, is abundant within presynaptic terminals. Altered SphK1/S1P metabolisms has been highlighted in many neurologic disorders with defective synaptic vesicle endocytosis. However, whether the SphK1/S1P axis may regulate synaptic vesicle endocytosis is unknown. Here, we identify that the SphK1/S1P axis regulates the kinetics of synaptic vesicle endocytosis in neurons, in addition to controlling fission-pore duration during single vesicle endocytosis in neuroendocrine chromaffin cells. The regulation of the SphK1/S1P axis in synaptic vesicle endocytosis is specific since it has a distinguished signaling pathway, which involves regulation of Ca2+ influx via TRPC5 channels. This discovery may provide novel mechanistic implications for the SphK1/S1P axis in brain functions under physiological and pathologic conditions.
Collapse
Affiliation(s)
- Zhong-Jiao Jiang
- Department of Biological Sciences, University of Illinois Chicago, Chicago, Illinois 60607
| | - Liang-Wei Gong
- Department of Biological Sciences, University of Illinois Chicago, Chicago, Illinois 60607
| |
Collapse
|
2
|
Spaulding SC, Bollag WB. The role of lipid second messengers in aldosterone synthesis and secretion. J Lipid Res 2022; 63:100191. [PMID: 35278411 PMCID: PMC9020094 DOI: 10.1016/j.jlr.2022.100191] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 11/23/2022] Open
Abstract
Second messengers are small rapidly diffusing molecules or ions that relay signals between receptors and effector proteins to produce a physiological effect. Lipid messengers constitute one of the four major classes of second messengers. The hydrolysis of two main classes of lipids, glycerophospholipids and sphingolipids, generate parallel profiles of lipid second messengers: phosphatidic acid (PA), diacylglycerol (DAG), and lysophosphatidic acid versus ceramide, ceramide-1-phosphate, sphingosine, and sphingosine-1-phosphate, respectively. In this review, we examine the mechanisms by which these lipid second messengers modulate aldosterone production at multiple levels. Aldosterone is a mineralocorticoid hormone responsible for maintaining fluid volume, electrolyte balance, and blood pressure homeostasis. Primary aldosteronism is a frequent endocrine cause of secondary hypertension. A thorough understanding of the signaling events regulating aldosterone biosynthesis may lead to the identification of novel therapeutic targets. The cumulative evidence in this literature emphasizes the critical roles of PA, DAG, and sphingolipid metabolites in aldosterone synthesis and secretion. However, it also highlights the gaps in our knowledge, such as the preference for phospholipase D-generated PA or DAG, as well as the need for further investigation to elucidate the precise mechanisms by which these lipid second messengers regulate optimal aldosterone production.
Collapse
Affiliation(s)
- Shinjini C Spaulding
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Wendy B Bollag
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA; Research Department, Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
3
|
Brown RDR, Veerman BEP, Oh J, Tate RJ, Torta F, Cunningham MR, Adams DR, Pyne S, Pyne NJ. A new model for regulation of sphingosine kinase 1 translocation to the plasma membrane in breast cancer cells. J Biol Chem 2021; 296:100674. [PMID: 33865856 PMCID: PMC8135045 DOI: 10.1016/j.jbc.2021.100674] [Citation(s) in RCA: 1] [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: 02/08/2021] [Revised: 04/01/2021] [Accepted: 04/14/2021] [Indexed: 01/24/2023] Open
Abstract
The translocation of sphingosine kinase 1 (SK1) to the plasma membrane (PM) is crucial in promoting oncogenesis. We have previously proposed that SK1 exists as both a monomer and dimer in equilibrium, although it is unclear whether these species translocate to the PM via the same or different mechanisms. We therefore investigated the structural determinants involved to better understand how translocation might potentially be targeted for therapeutic intervention. We report here that monomeric WT mouse SK1 (GFP-mSK1) translocates to the PM of MCF-7L cells stimulated with carbachol or phorbol 12-myristate 13-acetate, whereas the dimer translocates to the PM in response to sphingosine-1-phosphate; thus, the equilibrium between the monomer and dimer is sensitive to cellular stimulus. In addition, carbachol and phorbol 12-myristate 13-acetate induced translocation of monomeric GFP-mSK1 to lamellipodia, whereas sphingosine-1-phosphate induced translocation of dimeric GFP-mSK1 to filopodia, suggesting that SK1 regulates different cell biological processes dependent on dimerization. GFP-mSK1 mutants designed to modulate dimerization confirmed this difference in localization. Regulation by the C-terminal tail of SK1 was investigated using GFP-mSK1 truncations. Removal of the last five amino acids (PPEEP) prevented translocation of the enzyme to the PM, whereas removal of the last ten amino acids restored translocation. This suggests that the penultimate five amino acids (SRRGP) function as a translocation brake, which can be released by sequestration of the PPEEP sequence. We propose that these determinants alter the arrangement of N-terminal and C-terminal domains in SK1, leading to unique surfaces that promote differential translocation to the PM.
Collapse
Affiliation(s)
- Ryan D R Brown
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Ben E P Veerman
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Jeongah Oh
- SLING, Singapore Lipidomics Incubator, Life Sciences Institute and Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rothwelle J Tate
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Federico Torta
- SLING, Singapore Lipidomics Incubator, Life Sciences Institute and Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore, Singapore
| | - Margaret R Cunningham
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - David R Adams
- School of Engineering & Physical Sciences, Heriot-Watt University, Edinburgh, UK
| | - Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Nigel J Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK.
| |
Collapse
|
4
|
Dhangadamajhi G, Singh S. Malaria link of hypertension: a hidden syndicate of angiotensin II, bradykinin and sphingosine 1-phosphate. Hum Cell 2021; 34:734-744. [PMID: 33683655 DOI: 10.1007/s13577-021-00513-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/22/2021] [Indexed: 01/22/2023]
Abstract
In malaria-endemic countries, the burden of hypertension is on the rise. Although malaria and hypertension seem to have no direct link, several studies in recent years support their possible link. Three bioactive molecules such as angiotensin II (Ang II), bradykinin (BK) and sphingosine 1-phosphate (S1P) are crucial in regulating blood pressure. While the increased level of Ang II and S1P are responsible for inducing hypertension, BK is arthero-protective and anti-hypertensive. Therefore, in the present review, based on available literatures we highlight the present knowledge on the production and bioavailability of these molecules, the mechanism of their regulation of hypertension, and patho-physiological role in malaria. Further, a possible link between malaria and hypertension is hypothesized through various arguments based on experimental evidence. Understanding of their mechanisms of blood pressure regulation during malaria infection may open up avenues for drug therapeutics and management of malaria in co-morbidity with hypertension.
Collapse
Affiliation(s)
- Gunanidhi Dhangadamajhi
- Department of Biotechnology, Maharaja Sriramchandra Bhanjadeo University, Takatpur, Baripada, Odisha, 75003, India.
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
5
|
Role of bioactive sphingolipids in physiology and pathology. Essays Biochem 2021; 64:579-589. [PMID: 32579188 DOI: 10.1042/ebc20190091] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/27/2022]
Abstract
Sphingolipids are a class of complex lipids containing a backbone of sphingoid bases, namely the organic aliphatic amino alcohol sphingosine (Sph), that are essential constituents of eukaryotic cells. They were first described as major components of cell membrane architecture, but it is now well established that some sphingolipids are bioactive and can regulate key biological functions. These include cell growth and survival, cell differentiation, angiogenesis, autophagy, cell migration, or organogenesis. Furthermore, some bioactive sphingolipids are implicated in pathological processes including inflammation-associated illnesses such as atherosclerosis, rheumatoid arthritis, inflammatory bowel disease (namely Crohn's disease and ulcerative colitis), type II diabetes, obesity, and cancer. A major sphingolipid metabolite is ceramide, which is the core of sphingolipid metabolism and can act as second messenger, especially when it is produced at the plasma membrane of cells. Ceramides promote cell cycle arrest and apoptosis. However, ceramide 1-phosphate (C1P), the product of ceramide kinase (CerK), and Sph 1-phosphate (S1P), which is generated by the action of Sph kinases (SphK), stimulate cell proliferation and inhibit apoptosis. Recently, C1P has been implicated in the spontaneous migration of cells from some types of cancer, and can enhance cell migration/invasion of malignant cells through interaction with a Gi protein-coupled receptor. In addition, CerK and SphK are implicated in inflammatory responses, some of which are associated with cancer progression and metastasis. Hence, targeting these sphingolipid kinases to inhibit C1P or S1P production, or blockade of their receptors might contribute to the development of novel therapeutic strategies to reduce metabolic alterations and disease.
Collapse
|
6
|
Lee HY, Cho KM, Kim MK, Lee M, Kim H, Choi CY, Kim KK, Park JS, Kim HH, Bae YS. Sphingosylphosphorylcholine blocks ovariectomy-induced bone loss by suppressing Ca 2+ /calmodulin-mediated osteoclast differentiation. J Cell Mol Med 2020; 25:473-483. [PMID: 33230972 PMCID: PMC7810965 DOI: 10.1111/jcmm.16101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 09/29/2020] [Accepted: 11/01/2020] [Indexed: 12/19/2022] Open
Abstract
Osteoporosis is a disease in which bone mineral density decreases due to abnormal activity of osteoclasts, and is commonly found in post‐menopausal women who have decreased levels of female hormones. Sphingosylphosphorylcholine (SPC) is an important biological lipid that can be converted to sphingosine‐1‐phosphate (S1P) by autotaxin. S1P is known to be involved in osteoclast activation by stimulating osteoblasts, but bone regulation by SPC is not well understood. In this study, we found that SPC strongly inhibits RANKL‐induced osteoclast differentiation. SPC‐induced inhibitory effects on osteoclast differentiation were not affected by several antagonists of S1P receptors or pertussis toxin, suggesting cell surface receptor independency. However, SPC inhibited RANKL‐induced calcineurin activation and subsequent NFATc1 activity, leading to decrease of the expression of Trap and Ctsk. Moreover, we found that bone loss in an experimental osteoporosis mouse model was recovered by SPC injection. SPC also blocked ovariectomy‐induced body weight increase and Nfatc1 gene expression in mice. We also found that SPC inhibits RANKL‐induced osteoclast differentiation in human macrophages. Since currently available treatments for osteoporosis, such as administration of female hormones or hormone receptor modulators, show serious side effects, SPC has potential as a new agent for osteoporosis treatment.
Collapse
Affiliation(s)
- Ha Young Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Kwang Min Cho
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Min Kyung Kim
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Mingyu Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Hun Kim
- Department of Precision Medicine, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Cheol Yong Choi
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Joon Seong Park
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
7
|
Follicle-stimulating hormone promotes the proliferation of epithelial ovarian cancer cells by activating sphingosine kinase. Sci Rep 2020; 10:13834. [PMID: 32796926 PMCID: PMC7428003 DOI: 10.1038/s41598-020-70896-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 08/05/2020] [Indexed: 01/01/2023] Open
Abstract
Follicle-stimulating hormone (FSH) is closely related to the pathogenesis and progression of epithelial ovarian cancer (EOC). However, until now, knowledge relating to FSH-driven signalling pathways that lead to the growth of EOC remained incomplete. We sought to explore whether sphingosine kinase (SphK) could mediate FSH-induced ovarian cancer cell proliferation and which pathway might be involved in this process. The expression of phospho-SphK1 and phospho-SphK2 was detected in sections of EOC tissues by Immunohistochemical staining, and clinical significances were analyzed by statistical analysis. EOC cells were treated with FSH or/and SKI-II. CCK8 assays and colony formation assays were used to investigate cell proliferation. Western blot was carried out to detect protein expression in EOC cell line after treated with FSH. Here, for the first time, we provide evidence that high expression levels of phospho-SphK1 and phospho-SphK2 were both prognostic indicators of overall survival (OS) in EOC. Additionally, the expression levels of both phospho-SphK1 and phospho-SphK2 were closely correlated with the expression level of follicle-stimulating hormone receptor (FSHR) in ovarian cancer tissues. FSH stimulated the phosphorylation of both SphK1 and SphK2 and was able to regulate the survival and growth of ovarian cancer cells by activating SphK1 and SphK2 through ERK1/2. Both isoenzymes of SphK were equally responsible for FSH-induced cell proliferation of EOC. Both Erk1/2 and Akt activation play important roles in mediating FSH-induced cell proliferation after phosphorylation of SphK. Moreover, our data demonstrated that S1P receptor 1 (S1PR1) and S1PR3, key components of the SphK signalling system, were involved in FSH-mediated proliferation of EOC. Taken together, the results of the current study revealed that SphK is an essential mediator in FSH-induced proliferation of ovarian cancer cells in EOC, which indicates a new signalling pathway that controls FSH-mediated growth in EOC and suggests a new strategy that pharmaceutically targets both isoenzymes of SphK for the management of ovarian cancer.
Collapse
|
8
|
Jiang ZJ, Delaney TL, Zanin MP, Haberberger RV, Pitson SM, Huang J, Alford S, Cologna SM, Keating DJ, Gong LW. Extracellular and intracellular sphingosine-1-phosphate distinctly regulates exocytosis in chromaffin cells. J Neurochem 2019; 149:729-746. [PMID: 30963576 DOI: 10.1111/jnc.14703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/28/2018] [Accepted: 03/27/2019] [Indexed: 01/18/2023]
Abstract
Sphingosine-1-phosphate (S1P) is an essential bioactive sphingosine lipid involved in many neurological disorders. Sphingosine kinase 1 (SphK1), a key enzyme for S1P production, is concentrated in presynaptic terminals. However, the role of S1P/SphK1 signaling in exocytosis remains elusive. By detecting catecholamine release from single vesicles in chromaffin cells, we show that a dominant negative SphK1 (SphK1DN ) reduces the number of amperometric spikes and increases the duration of foot, which reflects release through a fusion pore, implying critical roles for S1P in regulating the rate of exocytosis and fusion pore expansion. Similar phenotypes were observed in chromaffin cells obtained from SphK1 knockout mice compared to those from wild-type mice. In addition, extracellular S1P treatment increased the number of amperometric spikes, and this increase, in turn, was inhibited by a selective S1P3 receptor blocker, suggesting extracellular S1P may regulate the rate of exocytosis via activation of S1P3. Furthermore, intracellular S1P application induced a decrease in foot duration of amperometric spikes in control cells, indicating intracellular S1P may regulate fusion pore expansion during exocytosis. Taken together, our study represents the first demonstration that S1P regulates exocytosis through distinct mechanisms: extracellular S1P may modulate the rate of exocytosis via activation of S1P receptors while intracellular S1P may directly control fusion pore expansion during exocytosis. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Zhong-Jiao Jiang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Taylor L Delaney
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mark P Zanin
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, P.R. China
| | - Rainer V Haberberger
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Jian Huang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Damien J Keating
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, Australia
| | - Liang-Wei Gong
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
9
|
Mamazhakypov A, Schermuly RT, Schaefer L, Wygrecka M. Lipids - two sides of the same coin in lung fibrosis. Cell Signal 2019; 60:65-80. [PMID: 30998969 DOI: 10.1016/j.cellsig.2019.04.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/07/2019] [Accepted: 04/12/2019] [Indexed: 12/16/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by progressive extracellular matrix deposition in the lung parenchyma leading to the destruction of lung structure, respiratory failure and premature death. Recent studies revealed that the pathogenesis of IPF is associated with alterations in the synthesis and the activity of lipids, lipid regulating proteins and cell membrane lipid transporters and receptors in different lung cells. Furthermore, deregulated lipid metabolism was found to contribute to the profibrotic phenotypes of lung fibroblasts and alveolar epithelial cells. Consequently, several pharmacological agents, targeting lipids, lipid mediators, and lipoprotein receptors, was successfully tested in the animal models of lung fibrosis and entered early phase clinical trials. In this review, we highlight new therapeutic options to counteract disturbed lipid hemostasis in the maladaptive lung remodeling.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany.
| | - Ralph T Schermuly
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany.
| | - Liliana Schaefer
- Goethe University School of Medicine, Frankfurt am Main, Germany.
| | - Malgorzata Wygrecka
- Department of Biochemistry, Universities of Giessen and Marburg Lung Center, Giessen, Germany.
| |
Collapse
|
10
|
Yue H, Hu B, Luo Z, Liu M. Metformin protects against sevoflurane-induced neuronal apoptosis through the S1P1 and ERK signaling pathways. Exp Ther Med 2019; 17:1463-1469. [PMID: 30680029 DOI: 10.3892/etm.2018.7098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/26/2018] [Indexed: 12/18/2022] Open
Abstract
The aim of the current study was to investigate whether metformin could counteract sevoflurane-induced neurotoxicity. In vitro experiments on the sevoflurane-induced nerve injury were performed using hippocampal neurons. Neuronal apoptosis was detected by an MTT assay. Protein expression levels of apoptosis-associated genes, including cleaved-caspase-3, apoptosis regulator BAX and apoptosis regulator Bcl-2 were detected by western blot analysis. The mechanism of the effect of metformin on sevoflurane-induced neuronal apoptosis was investigated using a sphingosine 1-phosphate receptor 1 (S1P1) antagonist (VPC23019) and mitogen-activated protein kinase kinase inhibitor (U0126). The current study revealed that metformin may reduce sevoflurane-induced neuronal apoptosis via activating mitogen-activated protein kinase (ERK)1/2 phosphorylation. VPC23019 and U0126 eliminated the neuroprotective effects of metformin on neuronal apoptosis, which suggests that metformin is able to protect against sevoflurane-induced neurotoxicity via activation of the S1P1-dependent ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Huiyu Yue
- Department of Anesthesiology, Shaan Xi Provincial Tumor Hospital, Xi'an, Shaanxi 710061, P.R. China
| | - Bin Hu
- Department of Anesthesiology, Yan'an University Affiliated Hospital, Yan'an, Shaanxi 716000, P.R. China
| | - Zhikai Luo
- Department of Anesthesiology, Yan'an University Affiliated Hospital, Yan'an, Shaanxi 716000, P.R. China
| | - Mei Liu
- Department of Anesthesiology, Yan'an University Affiliated Hospital, Yan'an, Shaanxi 716000, P.R. China
| |
Collapse
|
11
|
Werth S, Müller-Fielitz H, Raasch W. Obesity-stimulated aldosterone release is not related to an S1P-dependent mechanism. J Endocrinol 2017; 235:251-265. [PMID: 28970286 DOI: 10.1530/joe-16-0550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 09/26/2017] [Indexed: 11/08/2022]
Abstract
Aldosterone has been identified as an important factor in obesity-associated hypertension. Here, we investigated whether sphingosine-1-phosphate (S1P), which has previously been linked to obesity, increases aldosterone release. S1P-induced aldosterone release was determined in NCI H295R cells in the presence of S1P receptor (S1PR) antagonists. In vivo release of S1P (100-300 µg/kgbw) was investigated in pithed, lean Sprague Dawley (SD) rats, diet-obese spontaneous hypertensive rats (SHRs), as well as in lean or obese Zucker rats. Aldosterone secretion was increased in NCI H295R cells by S1P, the selective S1PR1 agonist SEW2871 and the selective S1PR2 antagonist JTE013. Treatment with the S1PR1 antagonist W146 or fingolimod and the S1PR1/3 antagonist VPbib2319 decreased baseline and/or S1P-stimulated aldosterone release. Compared to saline-treated SD rats, plasma aldosterone increased by ~50 pg/mL after infusing S1P. Baseline levels of S1P and aldosterone were higher in obese than in lean SHRs. Adrenal S1PR expression did not differ between chow- or CD-fed rats that had the highest S1PR1 and lowest S1PR4 levels. S1P induced a short-lasting increase in plasma aldosterone in obese, but not in lean SHRs. However, 2-ANOVA did not demonstrate any difference between lean and obese rats. S1P-induced aldosterone release was also similar between obese and lean Zucker rats. We conclude that S1P is a local regulator of aldosterone production. S1PR1 agonism induces an increase in aldosterone secretion, while stimulating adrenal S1PR2 receptor suppresses aldosterone production. A significant role of S1P in influencing aldosterone secretion in states of obesity seems unlikely.
Collapse
Affiliation(s)
- Stephan Werth
- Institute of Experimental and Clinical Pharmacology and ToxicologyUniversity of Lübeck, Lübeck, Germany
| | - Helge Müller-Fielitz
- Institute of Experimental and Clinical Pharmacology and ToxicologyUniversity of Lübeck, Lübeck, Germany
- CBBM (Center of Brain, Behavior and Metabolism)Lübeck, Germany
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and ToxicologyUniversity of Lübeck, Lübeck, Germany
- CBBM (Center of Brain, Behavior and Metabolism)Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research)partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| |
Collapse
|
12
|
Al Alam N, Kreydiyyeh SI. Signaling pathway involved in the inhibitory effect of FTY720P on the Na +/K + ATPase in HepG2 cells. J Cell Commun Signal 2017; 11:309-316. [PMID: 28197966 PMCID: PMC5704036 DOI: 10.1007/s12079-016-0369-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 11/28/2016] [Indexed: 02/05/2023] Open
Abstract
The Na+/K+ ATPase modulates the activity of many transporters in the liver, and maintains the ionic constancy of the intracellular milieu, preserving thus normal functioning of hepatocytes. Previous work showed that FTY720P, a sphingosine one phosphate receptor agonist used in the treatment of multiple sclerosis, exerts in HepG2 cells, an inhibitory effect on the activity of the ATPase, mediated via PGE2. This study is an attempt to identify the signaling molecules involved downstream of the prostaglandin. The activity of the ATPase was assayed by measuring the amount of inorganic phosphate liberated in presence and absence of ouabain, a specific inhibitor of the enzyme. The effect of FTY720P and PGE2 disappeared completely in presence of PF-04418948, a blocker of EP2 receptors, RpcAMP, an inhibitor of PKA, PD98059, an inhibitor of ERK, as well as in presence of PTIO, a nitric oxide synthase inhibitor, but was mimicked by butaprost, an EP2 agonist, dbcAMP, a cell permeable cAMP analogue, and SNAP1,a nitric oxide generator. PGE2 and dbcAMP increased the expression of phosphorylated ERK but not total ERK. This increase did not appear however in presence of PTIO, indicating that PKA is upstream of NO. It was concluded that FTY 720P induces PGE2 release which activates NOS leading to NO production and ERK activation. ERK then inhibits directly or indirectly the Na+/K+ ATPase.
Collapse
Affiliation(s)
- Nadine Al Alam
- Department of Biology, Faculty of Arts & Sciences, American University of Beirut, Beirut, Lebanon
| | | |
Collapse
|
13
|
Decoding resistant hypertension signalling pathways. Clin Sci (Lond) 2017; 131:2813-2834. [PMID: 29184046 DOI: 10.1042/cs20171398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/16/2017] [Accepted: 10/23/2017] [Indexed: 01/01/2023]
Abstract
Resistant hypertension (RH) is a clinical condition in which the hypertensive patient has become resistant to drug therapy and is often associated with increased cardiovascular morbidity and mortality. Several signalling pathways have been studied and related to the development and progression of RH: modulation of sympathetic activity by leptin and aldosterone, primary aldosteronism, arterial stiffness, endothelial dysfunction and variations in the renin-angiotensin-aldosterone system (RAAS). miRNAs comprise a family of small non-coding RNAs that participate in the regulation of gene expression at post-transcriptional level. miRNAs are involved in the development of both cardiovascular damage and hypertension. Little is known of the molecular mechanisms that lead to development and progression of this condition. This review aims to cover the potential roles of miRNAs in the mechanisms associated with the development and consequences of RH, and explore the current state of the art of diagnostic and therapeutic tools based on miRNA approaches.
Collapse
|
14
|
Vogt D, Stark H. Therapeutic Strategies and Pharmacological Tools Influencing S1P Signaling and Metabolism. Med Res Rev 2016; 37:3-51. [PMID: 27480072 DOI: 10.1002/med.21402] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 06/01/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023]
Abstract
During the last two decades the study of the sphingolipid anabolic, catabolic, and signaling pathways has attracted enormous interest. Especially the introduction of fingolimod into market as first p.o. therapeutic for the treatment of multiple sclerosis has boosted this effect. Although the complex regulation of sphingosine-1-phosphate (S1P) and other catabolic and anabolic sphingosine-related compounds is not fully understood, the influence on different (patho)physiological states from inflammation to cytotoxicity as well as the availability of versatile pharmacological tools that represent new approaches to study these states are described. Here, we have summarized various aspects concerning the many faces of sphingolipid function modulation by different pharmacological tools up to clinical candidates. Due to the immense heterogeneity of physiological or pharmacological actions and complex cross regulations, it is difficult to predict their role in upcoming therapeutic approaches. Currently, inflammatory, immunological, and/or antitumor aspects are discussed.
Collapse
Affiliation(s)
- Dominik Vogt
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, D-60438, Frankfurt, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225, Düsseldorf, Germany
| |
Collapse
|
15
|
Presa N, Gomez-Larrauri A, Rivera IG, Ordoñez M, Trueba M, Gomez-Muñoz A. Regulation of cell migration and inflammation by ceramide 1-phosphate. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:402-9. [DOI: 10.1016/j.bbalip.2016.02.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 02/05/2016] [Accepted: 02/08/2016] [Indexed: 12/13/2022]
|
16
|
Angiopoietin-like protein 8 (betatrophin) is a stress-response protein that down-regulates expression of adipocyte triglyceride lipase. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:130-137. [DOI: 10.1016/j.bbalip.2015.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/29/2015] [Accepted: 11/09/2015] [Indexed: 12/29/2022]
|
17
|
Abstract
Aldosterone is a steroid hormone synthesized in and secreted from the outer layer of the adrenal cortex, the zona glomerulosa. Aldosterone is responsible for regulating sodium homeostasis, thereby helping to control blood volume and blood pressure. Insufficient aldosterone secretion can lead to hypotension and circulatory shock, particularly in infancy. On the other hand, excessive aldosterone levels, or those too high for sodium status, can cause hypertension and exacerbate the effects of high blood pressure on multiple organs, contributing to renal disease, stroke, visual loss, and congestive heart failure. Aldosterone is also thought to directly induce end-organ damage, including in the kidneys and heart. Because of the significance of aldosterone to the physiology and pathophysiology of the cardiovascular system, it is important to understand the regulation of its biosynthesis and secretion from the adrenal cortex. Herein, the mechanisms regulating aldosterone production in zona glomerulosa cells are discussed, with a particular emphasis on signaling pathways involved in the secretory response to the main controllers of aldosterone production, the renin-angiotensin II system, serum potassium levels and adrenocorticotrophic hormone. The signaling pathways involved include phospholipase C-mediated phosphoinositide hydrolysis, inositol 1,4,5-trisphosphate, cytosolic calcium levels, calcium influx pathways, calcium/calmodulin-dependent protein kinases, diacylglycerol, protein kinases C and D, 12-hydroxyeicostetraenoic acid, phospholipase D, mitogen-activated protein kinase pathways, tyrosine kinases, adenylate cyclase, and cAMP-dependent protein kinase. A complete understanding of the signaling events regulating aldosterone biosynthesis may allow the identification of novel targets for therapeutic interventions in hypertension, primary aldosteronism, congestive heart failure, renal disease, and other cardiovascular disorders.
Collapse
Affiliation(s)
- Wendy B Bollag
- Charlie Norwood VA Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia at Georgia Regents University, Augusta, Georgia
| |
Collapse
|
18
|
Zhong Y, Wang K, Zhang X, Cai X, Chen Y, Deng Y. Nephrokeli, a Chinese herbal formula, may improve IgA nephropathy through regulation of the sphingosine-1-phosphate pathway. PLoS One 2015; 10:e0116873. [PMID: 25633986 PMCID: PMC4310606 DOI: 10.1371/journal.pone.0116873] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 12/15/2014] [Indexed: 01/12/2023] Open
Abstract
Nephrokeli (NPKL) is a Chinese herbal formula that has been used to treat patients with IgA nephropathy (IgAN) for improvement of proteinuria and kidney injury. However, the mechanism remains unclear. Sphingosine-1-phosphate (S1P) and its receptors S1PR2 and S1PR3 are known to play an important role in kidney disease. Here, we tested whether NPKL is able to regulate the S1P pathway in the kidney of IgAN rats. Four groups of rats were included in the study: Control, IgAN, IgAN treated with losartan, and IgAN treated with NPKL. The IgAN model was generated by injection of bovine serum albumin and staphylococcus enterotoxin B. We found that IgAN rats had increased staining for proliferating cell nuclear antigen (PCNA) in the mesangial area and increased mRNA and protein levels of S1PR2 and S1PR3 in the kidney compared to control rats. Connective tissue growth factor (CTGF), a downstream growth factor in the S1P pathway, was also elevated in the kidney of IgAN rats. Treatment with either NPKL or losartan was able to reduce PCNA staining and the expression of both S1PR2 and S1PR3 in the kidney of IgAN rats. However, NPKL (but not losartan treatment) reduced the expression of CTGF in the kidney of IgAN rats. In addition, we treated rat mesangial cells with sera collected from either NPKL-treated rats or control rats and found that NPKL-serum was able to reduce S1P-induced mesangial cell proliferation and the expression of S1PR2/S1PR3 and CTGF. NPKL also attenuates expression of fibrosis, inflammation, and oxidative stress markers in the kidney of IgAN rats. Our studies provide the mechanism by which NPKL attenuates kidney injury in IgAN rats.
Collapse
Affiliation(s)
- Yifei Zhong
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- * E-mail: (YZ); (YD)
| | - Ke Wang
- Surgical Research Institute of Traditional Chinese Medicine Combined with Western Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xianwen Zhang
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaofan Cai
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiping Chen
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yueyi Deng
- Department of Nephrology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- * E-mail: (YZ); (YD)
| |
Collapse
|
19
|
Langeslag M, Quarta S, Leitner MG, Kress M, Mair N. Sphingosine 1-phosphate to p38 signaling via S1P1 receptor and Gαi/o evokes augmentation of capsaicin-induced ionic currents in mouse sensory neurons. Mol Pain 2014; 10:74. [PMID: 25431213 PMCID: PMC4280769 DOI: 10.1186/1744-8069-10-74] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 11/03/2014] [Indexed: 12/13/2022] Open
Abstract
The perception of painful thermal stimuli by sensory neurons is largely mediated by TRPV1. Upon tissue injury or inflammation, S1P is secreted by thrombocytes as part of an inflammatory cocktail, which sensitizes nociceptive neurons towards thermal stimuli. S1P acts on G-protein coupled receptors that are expressed in sensory neurons and sensitize TRPV1 channels towards thermal stimuli. In this study, the S1P mediated signaling pathway required for sensitization of TRPV1 channels was explored.The capsaicin induced peak inward current (ICAPS) of sensory neurons was significantly increased after S1P stimulation within minutes after application. The potentiation of ICAPS resulted from activation of Gαi through G-protein coupled receptors for S1P. Consequently, Gαi led to a signaling cascade, involving phosphoinositide-3-kinase (PI3K) and protein kinase C, which augmented ICAPS in nociceptive neurons. The S1P1 receptor agonist SEW2871 resulted in activation of the same signaling pathway and potentiation of ICAPS. Furthermore, the mitogen-activated protein kinase p38 was phosphorylated after S1P stimulation and inhibition of p38 signaling by SB203580 prevented the S1P-induced ICAPS potentiation. The current data suggest that S1P sensitized ICAPS through G-protein coupled S1P1 receptor activation of Gαi-PI3K-PKC-p38 signaling pathway in sensory neurons.
Collapse
Affiliation(s)
- Michiel Langeslag
- />Division Physiology, DPMP, Medical University Innsbruck, Fritz-Pregl-Str. 3-I, 6020 Innsbruck, Austria
| | - Serena Quarta
- />Division Physiology, DPMP, Medical University Innsbruck, Fritz-Pregl-Str. 3-I, 6020 Innsbruck, Austria
| | - Michael G Leitner
- />Department of Neurophysiology, Institute for Physiology and Pathophysiology, Philipps University of Marburg, Marburg, Germany
| | - Michaela Kress
- />Division Physiology, DPMP, Medical University Innsbruck, Fritz-Pregl-Str. 3-I, 6020 Innsbruck, Austria
| | - Norbert Mair
- />Division Physiology, DPMP, Medical University Innsbruck, Fritz-Pregl-Str. 3-I, 6020 Innsbruck, Austria
| |
Collapse
|
20
|
Zhang W, Zhao J, Lee JF, Gartung A, Jawadi H, Lambiv WL, Honn KV, Lee MJ. ETS-1-mediated transcriptional up-regulation of CD44 is required for sphingosine-1-phosphate receptor subtype 3-stimulated chemotaxis. J Biol Chem 2013; 288:32126-32137. [PMID: 24064218 PMCID: PMC3820853 DOI: 10.1074/jbc.m113.495218] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/18/2013] [Indexed: 12/17/2022] Open
Abstract
Sphingosine-1-phosphate (S1P)-regulated chemotaxis plays critical roles in various physiological and pathophysiological conditions. S1P-regulated chemotaxis is mediated by the S1P family of G-protein-coupled receptors. However, molecular details of the S1P-regulated chemotaxis are incompletely understood. Cultured human lung adenocarcinoma cell lines abundantly express S1P receptor subtype 3 (S1P3), thus providing a tractable in vitro system to characterize molecular mechanism(s) underlying the S1P3 receptor-regulated chemotactic response. S1P treatment enhances CD44 expression and induces membrane localization of CD44 polypeptides via the S1P3/Rho kinase (ROCK) signaling pathway. Knockdown of CD44 completely diminishes the S1P-stimulated chemotaxis. Promoter analysis suggests that the CD44 promoter contains binding sites of the ETS-1 (v-ets erythroblastosis virus E26 oncogene homolog 1) transcriptional factor. ChIP assay confirms that S1P treatment stimulates the binding of ETS-1 to the CD44 promoter region. Moreover, S1P induces the expression and nuclear translocation of ETS-1. Knockdown of S1P3 or inhibition of ROCK abrogates the S1P-induced ETS-1 expression. Furthermore, knockdown of ETS-1 inhibits the S1P-induced CD44 expression and cell migration. In addition, we showed that S1P3/ROCK signaling up-regulates ETS-1 via the activity of JNK. Collectively, we characterized a novel signaling axis, i.e., ROCK-JNK-ETS-1-CD44 pathway, which plays an essential role in the S1P3-regulated chemotactic response.
Collapse
Affiliation(s)
- Wenliang Zhang
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | - Jiawei Zhao
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | - Jen-Fu Lee
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | - Allison Gartung
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | | | | | - Kenneth V Honn
- From the Department of Pathology,; the Bioactive Lipid Research Program,; the Karmanos Cancer Institute
| | - Menq-Jer Lee
- From the Department of Pathology,; the Bioactive Lipid Research Program,; the Karmanos Cancer Institute; the Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan 48201.
| |
Collapse
|
21
|
Sewer MB, Li D. Regulation of adrenocortical steroid hormone production by RhoA-diaphanous 1 signaling and the cytoskeleton. Mol Cell Endocrinol 2013; 371. [PMID: 23186810 PMCID: PMC3926866 DOI: 10.1016/j.mce.2012.11.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The production of glucocorticoids and aldosterone in the adrenal cortex is regulated at multiple levels. Biosynthesis of these hormones is initiated when cholesterol, the substrate, enters the inner mitochondrial membrane for conversion to pregnenolone. Unlike most metabolic pathways, the biosynthesis of adrenocortical steroid hormones is unique because some of the enzymes are localized in mitochondria and others in the endoplasmic reticulum (ER). Although much is known about the factors that control the transcription and activities of the proteins that are required for steroid hormone production, the parameters that govern the exchange of substrates between the ER and mitochondria are less well understood. This short review summarizes studies that have begun to provide insight into the role of the cytoskeleton, mitochondrial transport, and the physical interaction of the ER and mitochondria in the production of adrenocortical steroid hormones.
Collapse
Affiliation(s)
- Marion B Sewer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093-0704, USA.
| | | |
Collapse
|
22
|
The PI3K/AKT/mTOR signaling pathway is overactivated in primary aldosteronism. PLoS One 2013; 8:e62399. [PMID: 23626817 PMCID: PMC3633845 DOI: 10.1371/journal.pone.0062399] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 03/20/2013] [Indexed: 12/21/2022] Open
Abstract
Background To date, the available non-invasive remedies for primary aldosteronism are not satisfactory in clinical practice. The phosphoinositide 3-kinase (PI3Ks)/protein kinase B (PKB or AKT)/mammalian target of rapamycin (mTOR) signaling pathway is essential for tumorigenesis and metastasis in many types of human tumors, including renal cancer, adrenal carcinoma and pheochromocytoma. The possibility that this pathway is also necessary for the pathogenesis of primary aldosteronism has not yet been explored. To answer this question, we investigated the activity of the PI3K/AKT/mTOR signaling pathway in normal adrenal glands (NAGs), primary aldosteronism (PA) patients and NCI-H295R cells. Methodology/Principal Findings Between January 2005 and December 2011, we retrospectively reviewed the records of 45 patients with PA. We compared clinical characteristics (age, gender and biochemical data) and the expression of phospho-AKT (p-AKT), phospho-mTOR (p-mTOR), phospho-S6 (p-S6) and vascular endothelial growth factor (VEGF) by immunohistochemical staining and western blotting, analyzing 30 aldosterone-producing adenomas (APAs), 15 idiopathic hyperaldosteronism (IHA) tissues and 12 NAGs following nephrectomy for renal tumors (control group). Compared with the control group, most of the PA patients presented with polydipsia, polyuria, resistant hypertension, profound hypokalemia, hyperaldosteronemia and decreased plasma renin activity. Compared with normal zona glomerulosa, the levels of p-AKT, p-mTOR, p-S6 and VEGF were significantly upregulated in APA and IHA. No significant differences were found between APA and IHA in the expression of these proteins. Additionally, positive correlations existed between the plasma aldosterone levels and the expression of p-AKT and p-mTOR. In vitro studies showed that mTOR inhibitor rapamycin could inhibit cell proliferation in NCI-H295R cells in a dose- and time-dependent manner. Furthermore, this inhibitor also decreased aldosterone secretion. Conclusions Our data suggest that the PI3K/AKT/mTOR signaling pathway, which was overactivated in APA and IHA compared with normal zona glomerulosa, may mediate aldosterone hypersecretion and participate in the development of PA.
Collapse
|
23
|
Phospholipase D1 mediates bFGF-induced Bcl-2 expression leading to neurite outgrowth in H19-7 cells. Biochem J 2012; 441:407-16. [PMID: 21916846 DOI: 10.1042/bj20110302] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The purpose of the present study was to investigate the role of PLD (phospholipase D) in bFGF (basic fibroblast growth factor)-induced Bcl-2 expression and to examine whether overexpressed Bcl-2 influences neurite outgrowth in immortalized hippocampal progenitor cells (H19-7 cells). We found that Bcl-2 expression was maximally induced by bFGF within 24 h, and that this effect was reduced by inhibiting PLD1 expression with PLD1 small interfering RNA or by overexpressing DN (dominant-negative)-PLD1, whereas PLD1 overexpression markedly induced Bcl-2 expression. bFGF treatment activated Ras, Src, PI3K (phosphoinositide 3-kinase), PLCγ (phospholipase Cγ) and PKCα (protein kinase Cα). Among these molecules, Src and PKCα were not required for Bcl-2 expression. PLD activity was decreased by Ras, PI3K or PLCγ inhibitor, suggesting that PLD1 activation occurred through Ras, PI3K or PLCγ. We found that Ras was the most upstream molecule among these proteins, followed by the PI3K/PLCγ pathway, indicating that bFGF-induced PLD activation took place through the Ras/PI3K/PLCγ pathway. Furthermore, PLD1 was required for activation of JNK (c-Jun N-terminal kinase), which led to activation of STAT3 (signal transducer and activator of transcription 3) and finally Bcl-2 expression. When Bcl-2 was overexpressed, neurite outgrowth was stimulated along with induction of neurotrophic factors such as brain-derived neurotrophic factor and neurotrophin 4/5. In conclusion, PLD1 acts as a downstream effector of bFGF/Ras/PI3K/PLCγ signalling and regulates Bcl-2 expression through JNK/STAT3, which leads to neurite outgrowth in H19-7 cells.
Collapse
|
24
|
Lucki NC, Bandyopadhyay S, Wang E, Merrill AH, Sewer MB. Acid ceramidase (ASAH1) is a global regulator of steroidogenic capacity and adrenocortical gene expression. Mol Endocrinol 2012; 26:228-43. [PMID: 22261821 PMCID: PMC3275158 DOI: 10.1210/me.2011-1150] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 12/14/2011] [Indexed: 11/19/2022] Open
Abstract
In H295R human adrenocortical cells, ACTH rapidly activates ceramide (Cer) and sphingosine (SPH) turnover with a concomitant increase in SPH-1-phosphate secretion. These bioactive lipids modulate adrenocortical steroidogenesis, primarily by acting as second messengers in the protein kinase A/cAMP-dependent pathway. Acid ceramidase (ASAH1) directly regulates the intracellular balance of Cer, SPH, and SPH-1-phosphate by catalyzing the hydrolysis of Cer into SPH. ACTH/cAMP signaling stimulates ASAH1 transcription and activity, supporting a role for this enzyme in glucocorticoid production. Here, the role of ASAH1 in regulating steroidogenic capacity was examined using a tetracycline-inducible ASAH1 short hairpin RNA H295R human adrenocortical stable cell line. We show that ASAH1 suppression increases the transcription of multiple steroidogenic genes, including Cytochrome P450 monooxygenase (CYP)17A1, CYP11B1/2, CYP21A2, steroidogenic acute regulatory protein, hormone-sensitive lipase, 18-kDa translocator protein, and the melanocortin-2 receptor. Induced gene expression positively correlated with enhanced histone H3 acetylation at target promoters. Repression of ASAH1 expression also induced the expression of members of the nuclear receptor nuclear receptor subfamily 4 (NR4A) family while concomitantly suppressing the expression of dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1. ASAH1 knockdown altered the expression of genes involved in sphingolipid metabolism and changed the cellular amounts of distinct sphingolipid species. Finally, ASAH1 silencing increased basal and cAMP-dependent cortisol and dehydroepiandrosterone secretion, establishing ASAH1 as a pivotal regulator of steroidogenic capacity in the human adrenal cortex.
Collapse
Affiliation(s)
- Natasha C Lucki
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia 30332-0230, USA
| | | | | | | | | |
Collapse
|
25
|
Kolesnikov YS, Nokhrina KP, Kretynin SV, Volotovski ID, Martinec J, Romanov GA, Kravets VS. Molecular structure of phospholipase D and regulatory mechanisms of its activity in plant and animal cells. BIOCHEMISTRY (MOSCOW) 2012; 77:1-14. [DOI: 10.1134/s0006297912010014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Lucki NC, Li D, Sewer MB. Sphingosine-1-phosphate rapidly increases cortisol biosynthesis and the expression of genes involved in cholesterol uptake and transport in H295R adrenocortical cells. Mol Cell Endocrinol 2012; 348:165-75. [PMID: 21864647 PMCID: PMC3508734 DOI: 10.1016/j.mce.2011.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 07/26/2011] [Accepted: 08/03/2011] [Indexed: 12/22/2022]
Abstract
In the acute phase of adrenocortical steroidogenesis, adrenocorticotrophin (ACTH) activates a cAMP/PKA-signaling pathway that promotes the transport of free cholesterol to the inner mitochondrial membrane. We have previously shown that ACTH rapidly stimulates the metabolism of sphingolipids and the secretion of sphingosine-1-phosphate (S1P) in H295R cells. In this study, we examined the effect of S1P on genes involved in the acute phase of steroidogenesis. We show that S1P increases the expression of steroidogenic acute regulatory protein (StAR), 18-kDa translocator protein (TSPO), low-density lipoprotein receptor (LDLR), and scavenger receptor class B type I (SR-BI). S1P-induced StAR mRNA expression requires Gα(i) signaling, phospholipase C (PLC), Ca(2+)/calmodulin-dependent kinase II (CamKII), and ERK1/2 activation. S1P also increases intracellular Ca(2+), the phosphorylation of hormone sensitive lipase (HSL) at Ser(563), and cortisol secretion. Collectively, these findings identify multiple roles for S1P in the regulation of glucocorticoid biosynthesis.
Collapse
Affiliation(s)
- Natasha C. Lucki
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332-0230
| | - Donghui Li
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093-0704
| | - Marion B. Sewer
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093-0704
| |
Collapse
|
27
|
Lucki NC, Sewer MB. The interplay between bioactive sphingolipids and steroid hormones. Steroids 2010; 75:390-9. [PMID: 20138078 PMCID: PMC2854287 DOI: 10.1016/j.steroids.2010.01.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2009] [Revised: 01/25/2010] [Accepted: 01/26/2010] [Indexed: 01/02/2023]
Abstract
Steroid hormones regulate various physiological processes including development, reproduction, and metabolism. These regulatory molecules are synthesized from cholesterol in endocrine organs - such as the adrenal glands and gonads - via a multi-step enzymatic process that is catalyzed by the cytochrome P450 superfamily of monooxygenases and hydroxysteroid dehydrogenases. Steroidogenesis is induced by trophic peptide hormones primarily via the activation of a cAMP/protein kinase A (PKA)-dependent pathway. However, other signaling molecules, including cytokines and growth factors, control the steroid hormone biosynthetic pathway. More recently, sphingolipids, including ceramide, sphingosine-1-phosphate, and sphingosine, have been found to modulate steroid hormone secretion at multiple levels. In this review, we provide a brief overview of the mechanisms by which sphingolipids regulate steroidogenesis. In addition, we discuss how steroid hormones control sphingolipid metabolism. Finally, we outline evidence supporting the emerging role of bioactive sphingolipids in various nuclear processes and discuss a role for nuclear sphingolipid metabolism in the control of gene transcription.
Collapse
Affiliation(s)
- Natasha C. Lucki
- School of Biology and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 310 Ferst Dr., Atlanta, GA 30332
| | - Marion B. Sewer
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Dr. MC0704, La Jolla, CA 92093
| |
Collapse
|
28
|
Rotstein NP, Miranda GE, Abrahan CE, German OL. Regulating survival and development in the retina: key roles for simple sphingolipids. J Lipid Res 2010; 51:1247-62. [PMID: 20100817 PMCID: PMC3035489 DOI: 10.1194/jlr.r003442] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 01/25/2010] [Indexed: 12/28/2022] Open
Abstract
Many sphingolipids have key functions in the regulation of crucial cellular processes. Ceramide (Cer) and sphingosine (Sph) induce growth arrest and cell death in multiple situations of cellular stress. On the contrary, sphingosine-1-phosphate (S1P), the product of Sph phosphorylation, promotes proliferation, differentiation, and survival in different cell systems. This review summarizes the roles of these simple sphingolipids in different tissues and then analyzes their possible functions in the retina. Alterations in proliferation, neovascularization, differentiation, and cell death are critical in major retina diseases and collective evidence points to a role for sphingolipids in these processes. Cer induces inflammation and apoptosis in endothelial and retinal pigmented epithelium cells, leading to several retinopathies. S1P can prevent this death but also promotes cell proliferation that might lead to neovascularization and fibrosis. Recent data support Cer and Sph as crucial mediators in the induction of photoreceptor apoptosis in diverse models of oxidative damage and neurodegeneration, and suggest that regulating their metabolism can prevent this death. New evidence proposes a central role for S1P controlling photoreceptor survival and differentiation. Finally, this review discusses the ability of trophic factors to regulate sphingolipid metabolism and transactivate S1P signaling pathways to control survival and development in retina photoreceptors.
Collapse
Affiliation(s)
- Nora P Rotstein
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-CONICET, Bahía Blanca, Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
29
|
Williams TA, Monticone S, Morello F, Liew CC, Mengozzi G, Pilon C, Asioli S, Sapino A, Veglio F, Mulatero P. Teratocarcinoma-Derived Growth Factor-1 Is Upregulated in Aldosterone-Producing Adenomas and Increases Aldosterone Secretion and Inhibits Apoptosis In Vitro. Hypertension 2010; 55:1468-75. [DOI: 10.1161/hypertensionaha.110.150318] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Tracy A. Williams
- From the Division of Internal Medicine and Hypertension Unit, Department of Medicine and Experimental Oncology (T.A.W., S.M., F.M., F.V., P.M.) and Department of Biomedical Sciences and Human Oncology (S.A., A.S.), University of Torino, Torino, Italy; GeneNews Ltd (C.-C.L.), Richmond Hill, Ontario, Canada; Clinical Chemistry Laboratory (G.M.), AOU San Giovanni Battista, Torino, Italy; Department of Medical and Surgical Sciences (C.P.), University of Padova, Padova, Italy
| | - Silvia Monticone
- From the Division of Internal Medicine and Hypertension Unit, Department of Medicine and Experimental Oncology (T.A.W., S.M., F.M., F.V., P.M.) and Department of Biomedical Sciences and Human Oncology (S.A., A.S.), University of Torino, Torino, Italy; GeneNews Ltd (C.-C.L.), Richmond Hill, Ontario, Canada; Clinical Chemistry Laboratory (G.M.), AOU San Giovanni Battista, Torino, Italy; Department of Medical and Surgical Sciences (C.P.), University of Padova, Padova, Italy
| | - Fulvio Morello
- From the Division of Internal Medicine and Hypertension Unit, Department of Medicine and Experimental Oncology (T.A.W., S.M., F.M., F.V., P.M.) and Department of Biomedical Sciences and Human Oncology (S.A., A.S.), University of Torino, Torino, Italy; GeneNews Ltd (C.-C.L.), Richmond Hill, Ontario, Canada; Clinical Chemistry Laboratory (G.M.), AOU San Giovanni Battista, Torino, Italy; Department of Medical and Surgical Sciences (C.P.), University of Padova, Padova, Italy
| | - Choong-Chin Liew
- From the Division of Internal Medicine and Hypertension Unit, Department of Medicine and Experimental Oncology (T.A.W., S.M., F.M., F.V., P.M.) and Department of Biomedical Sciences and Human Oncology (S.A., A.S.), University of Torino, Torino, Italy; GeneNews Ltd (C.-C.L.), Richmond Hill, Ontario, Canada; Clinical Chemistry Laboratory (G.M.), AOU San Giovanni Battista, Torino, Italy; Department of Medical and Surgical Sciences (C.P.), University of Padova, Padova, Italy
| | - Giulio Mengozzi
- From the Division of Internal Medicine and Hypertension Unit, Department of Medicine and Experimental Oncology (T.A.W., S.M., F.M., F.V., P.M.) and Department of Biomedical Sciences and Human Oncology (S.A., A.S.), University of Torino, Torino, Italy; GeneNews Ltd (C.-C.L.), Richmond Hill, Ontario, Canada; Clinical Chemistry Laboratory (G.M.), AOU San Giovanni Battista, Torino, Italy; Department of Medical and Surgical Sciences (C.P.), University of Padova, Padova, Italy
| | - Catia Pilon
- From the Division of Internal Medicine and Hypertension Unit, Department of Medicine and Experimental Oncology (T.A.W., S.M., F.M., F.V., P.M.) and Department of Biomedical Sciences and Human Oncology (S.A., A.S.), University of Torino, Torino, Italy; GeneNews Ltd (C.-C.L.), Richmond Hill, Ontario, Canada; Clinical Chemistry Laboratory (G.M.), AOU San Giovanni Battista, Torino, Italy; Department of Medical and Surgical Sciences (C.P.), University of Padova, Padova, Italy
| | - Sofia Asioli
- From the Division of Internal Medicine and Hypertension Unit, Department of Medicine and Experimental Oncology (T.A.W., S.M., F.M., F.V., P.M.) and Department of Biomedical Sciences and Human Oncology (S.A., A.S.), University of Torino, Torino, Italy; GeneNews Ltd (C.-C.L.), Richmond Hill, Ontario, Canada; Clinical Chemistry Laboratory (G.M.), AOU San Giovanni Battista, Torino, Italy; Department of Medical and Surgical Sciences (C.P.), University of Padova, Padova, Italy
| | - Anna Sapino
- From the Division of Internal Medicine and Hypertension Unit, Department of Medicine and Experimental Oncology (T.A.W., S.M., F.M., F.V., P.M.) and Department of Biomedical Sciences and Human Oncology (S.A., A.S.), University of Torino, Torino, Italy; GeneNews Ltd (C.-C.L.), Richmond Hill, Ontario, Canada; Clinical Chemistry Laboratory (G.M.), AOU San Giovanni Battista, Torino, Italy; Department of Medical and Surgical Sciences (C.P.), University of Padova, Padova, Italy
| | - Franco Veglio
- From the Division of Internal Medicine and Hypertension Unit, Department of Medicine and Experimental Oncology (T.A.W., S.M., F.M., F.V., P.M.) and Department of Biomedical Sciences and Human Oncology (S.A., A.S.), University of Torino, Torino, Italy; GeneNews Ltd (C.-C.L.), Richmond Hill, Ontario, Canada; Clinical Chemistry Laboratory (G.M.), AOU San Giovanni Battista, Torino, Italy; Department of Medical and Surgical Sciences (C.P.), University of Padova, Padova, Italy
| | - Paolo Mulatero
- From the Division of Internal Medicine and Hypertension Unit, Department of Medicine and Experimental Oncology (T.A.W., S.M., F.M., F.V., P.M.) and Department of Biomedical Sciences and Human Oncology (S.A., A.S.), University of Torino, Torino, Italy; GeneNews Ltd (C.-C.L.), Richmond Hill, Ontario, Canada; Clinical Chemistry Laboratory (G.M.), AOU San Giovanni Battista, Torino, Italy; Department of Medical and Surgical Sciences (C.P.), University of Padova, Padova, Italy
| |
Collapse
|
30
|
Donovan EE, Pelanda R, Torres RM. S1P3 confers differential S1P-induced migration by autoreactive and non-autoreactive immature B cells and is required for normal B-cell development. Eur J Immunol 2010; 40:688-98. [PMID: 20039302 DOI: 10.1002/eji.200939858] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
During B-cell development, immature B-cell fate is determined by whether the BCR is engaged in the bone marrow. Immature B cells that are non-autoreactive continue maturation and emigrate from the marrow, whereas autoreactive immature B cells remain and are tolerized. However, the microenvironment where these events occur and the chemoattractants responsible for immature B-cell trafficking within and out of the bone marrow remain largely undefined. Sphingosine 1-phosphate (S1P) is a chemoattractant that directs lymphocyte trafficking and thymocyte egress and in this study we investigated whether S1P contributes to B-cell development, egress and positioning within the bone marrow. Our findings show that immature B cells are chemotactic toward S1P but that this response is dependent on Ag receptor specificity: non-autoreactive, but not autoreactive, immature B cells migrate toward S1P and are shown to require S1P3 receptor for this response. Despite this response, S1P3 is shown not to facilitate immature B-cell egress but is required for normal B-cell development including the positioning of transitional B cells within bone marrow sinusoids. These data indicate that S1P3 signaling directs immature B cells to a bone marrow microenvironment important for both tolerance induction and maturation.
Collapse
Affiliation(s)
- Erin E Donovan
- Integrated Department of Immunology, National Jewish Health, University of Colorado Denver, Denver, CO, USA
| | | | | |
Collapse
|
31
|
Gangoiti P, Camacho L, Arana L, Ouro A, Granado MH, Brizuela L, Casas J, Fabriás G, Abad JL, Delgado A, Gómez-Muñoz A. Control of metabolism and signaling of simple bioactive sphingolipids: Implications in disease. Prog Lipid Res 2010; 49:316-34. [PMID: 20193711 DOI: 10.1016/j.plipres.2010.02.004] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 02/18/2010] [Accepted: 02/22/2010] [Indexed: 01/05/2023]
Abstract
Simple bioactive sphingolipids include ceramide, sphingosine and their phosphorylated forms sphingosine 1-phosphate and ceramide 1-phosphate. These molecules are crucial regulators of cell functions. In particular, they play important roles in the regulation of angiogenesis, apoptosis, cell proliferation, differentiation, migration, and inflammation. Decoding the mechanisms by which these cellular functions are regulated requires detailed understanding of the signaling pathways that are implicated in these processes. Most importantly, the development of inhibitors of the enzymes involved in their metabolism may be crucial for establishing new therapeutic strategies for treatment of disease.
Collapse
Affiliation(s)
- Patricia Gangoiti
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
García-Pacios M, Collado MI, Busto JV, Sot J, Alonso A, Arrondo JLR, Goñi FM. Sphingosine-1-phosphate as an amphipathic metabolite: its properties in aqueous and membrane environments. Biophys J 2009; 97:1398-407. [PMID: 19720028 DOI: 10.1016/j.bpj.2009.07.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 07/01/2009] [Accepted: 07/01/2009] [Indexed: 12/22/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is currently considered to be an important signaling molecule in cell metabolism. We studied a number of relevant biophysical properties of S1P, using mainly Langmuir balance, differential scanning calorimetry, (31)P-NMR, and infrared (IR) spectroscopy. We found that, at variance with other, structurally related sphingolipids that are very hydrophobic, S1P may occur in either an aqueous dispersion or a bilayer environment. S1P behaves in aqueous media as a soluble amphiphile, with a critical micelle concentration of approximately 12 muM. Micelles give rise to larger aggregates (in the micrometer size range) at and above a 1 mM concentration. The aggregates display a thermotropic transition at approximately 60 degrees C, presumably due to the formation of smaller structures at the higher temperatures. S1P can also be studied in mixtures with phospholipids. Studies with dielaidoylphosphatidylethanolamine (DEPE) or deuterated dipalmitoylphosphatidylcholine (DPPC) show that S1P modifies the gel-fluid transition of the glycerophospholipids, shifting it to lower temperatures and decreasing the transition enthalpy. Low (<10 mol %) concentrations of S1P also have a clear effect on the lamellar-to-inverted hexagonal transition of DEPE, i.e., they increase the transition temperature and stabilize the lamellar versus the inverted hexagonal phase. IR spectroscopy of natural S1P mixed with deuterated DPPC allows the independent observation of transitions in each molecule, and demonstrates the existence of molecular interactions between S1P and the phospholipid at the polar headgroup level that lead to increased hydration of the carbonyl group. The combination of calorimetric, IR, and NMR data allowed the construction of a temperature-composition diagram ("partial phase diagram") to facilitate a comparative study of the properties of S1P and other related lipids (ceramide and sphingosine) in membranes. In conclusion, two important differences between S1P and ceramide are that S1P stabilizes the lipid bilayer structure, and physiologically relevant concentrations of S1P can exist dispersed in the cytosol.
Collapse
|
33
|
Lucki N, Sewer MB. The cAMP-responsive element binding protein (CREB) regulates the expression of acid ceramidase (ASAH1) in H295R human adrenocortical cells. Biochim Biophys Acta Mol Cell Biol Lipids 2009; 1791:706-13. [PMID: 19298866 DOI: 10.1016/j.bbalip.2009.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 02/19/2009] [Accepted: 03/06/2009] [Indexed: 01/09/2023]
Abstract
Acid ceramidase (encoded by ASAH1) is a lipid hydrolase that catalyzes the conversion of ceramide (cer) into sphingosine (SPH) and a free fatty acid. Adrenocortical steroidogenesis is regulated by the trophic peptide hormone adrenocorticotropin (ACTH), which induces the expression of steroidogenic genes in the human adrenal cortex primarily via a cAMP/protein kinase A (PKA)-dependent pathway. ACTH also stimulates sphingolipid metabolism in H295R adrenocortical cells leading to changes in steroidogenic gene expression. Based on our previous data identifying SPH as an antagonist for the nuclear receptor steroidogenic factor 1 (SF-1) and the role of ACTH-stimulated changes in sphingolipid metabolism on steroidogenic gene transcription, the aim of the current study was to determine the role of ACTH signaling in regulating the expression of the ASAH1 gene in H295R cells. We show that activation of the ACTH signaling pathway induces ASAH1 gene expression by stimulating the binding of the cAMP-responsive element binding protein (CREB) to multiple regions of the ASAH1 promoter. CREB binding promotes the recruitment of the coactivators CREB binding protein (CBP) and p300 to the CREB-responsive regions of the promoter. Consistent with transcriptional activation, we show that cAMP signaling increases the trimethylation of Lys 4 on histone H3 (H3K4) along the ASAH1 promoter. Finally, RNA interference (RNAi) experiments demonstrate that CREB is indispensable for cAMP-induced ASAH1 transcription. These data identify the ACTH/cAMP signaling pathway and CREB as transcriptional regulators of the ASAH1 gene in the human adrenal cortex.
Collapse
Affiliation(s)
- Natasha Lucki
- Parker H. Petit Institute for Bioengineering and Bioscience, Atlanta, GA 30332-0230, USA
| | | |
Collapse
|
34
|
Ansurudeen I, Willenberg HS, Kopprasch S, Krug AW, Ehrhart-Bornstein M, Bornstein SR. Endothelial factors mediate aldosterone release via PKA-independent pathways. Mol Cell Endocrinol 2009; 300:66-70. [PMID: 19073232 DOI: 10.1016/j.mce.2008.11.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2008] [Revised: 11/06/2008] [Accepted: 11/16/2008] [Indexed: 12/15/2022]
Abstract
Aldosterone synthesis is primarily regulated by angiotensin II and potassium ions. In addition, endothelial cell-secreted factors have been shown to regulate mineralocorticoid release. We analyzed the pathways that mediate endothelial cell-factor-induced aldosterone release from adrenocortical cells, NCI-H295R using endothelial cell-conditioned medium (ECM). The cAMP antagonist Rp-cAMP caused a 44% decrease in the ECM-induced aldosterone release but inhibition of cAMP-dependent PKA had no effect on aldosterone release. Interestingly, inhibition of cAMP-regulated guanine nucleotide exchange factor Epac with brefeldin-A decreased the ECM-induced aldosterone release by 45%. Similarly, inhibition of p38 MAP-kinase; PI-3-kinase and PKB significantly reduced the ECM-induced aldosterone release whereas inhibition of ERK1/2 and PKC did not decrease aldosterone release. These results provide evidence for the existence of a cAMP-dependent but PKA-independent pathway in mediating the ECM-induced aldosterone release and the significant influence of more than one signaling mechanism.
Collapse
Affiliation(s)
- Ishrath Ansurudeen
- Department of Medicine III, Carl Gustav Carus Medical School, University of Technology, Fetscherstrasse 74, 01307 Dresden, Germany.
| | | | | | | | | | | |
Collapse
|
35
|
Greenspon J, Li R, Xiao L, Rao JN, Marasa BS, Strauch ED, Wang JY, Turner DJ. Sphingosine-1-phosphate protects intestinal epithelial cells from apoptosis through the Akt signaling pathway. Dig Dis Sci 2009; 54:499-510. [PMID: 18654850 PMCID: PMC2696985 DOI: 10.1007/s10620-008-0393-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2008] [Accepted: 06/17/2008] [Indexed: 12/09/2022]
Abstract
OBJECTIVE The regulation of apoptosis of intestinal mucosal cells is important in maintenance of normal intestinal physiology. SUMMARY Sphingosine-1-phosphate (S1P) has been shown to play a critical role in cellular protection to otherwise lethal stimuli in several nonintestinal tissues. METHODS The current study determines whether S1P protected normal intestinal epithelial cells (IECs) from apoptosis and whether Akt activation was the central pathway for this effect. RESULTS S1P demonstrated significantly reduced levels of apoptosis induced by tumor necrosis factor-alpha (TNF-alpha)/cycloheximide (CHX). S1P induced increased levels of phosphorylated Akt and increased Akt activity, but did not affect total amounts of Akt. This activation of Akt was associated with decreased levels of both caspase-3 protein levels and of caspase-3 activity. Inactivation of Akt by treatment with the PI3K chemical inhibitor LY294002 or by overexpression of the dominant negative mutant of Akt (DNMAkt) prevented the protective effect of S1P on apoptosis. Additionally, silencing of the S1P-1 receptor by specific siRNA demonstrated a lesser decrease in apoptosis to S1P exposure. CONCLUSION These results indicate that S1P protects intestinal epithelial cells from apoptosis via an Akt-dependent pathway.
Collapse
Affiliation(s)
- Jose Greenspon
- Department of Surgery, Cell Biology Group, Baltimore Veterans Affairs Medical Center, University of Maryland School of Medicine, 10 N. Greene Street, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Sewer MB, Li D. Regulation of steroid hormone biosynthesis by the cytoskeleton. Lipids 2008; 43:1109-15. [PMID: 18726632 PMCID: PMC2717900 DOI: 10.1007/s11745-008-3221-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2008] [Accepted: 07/31/2008] [Indexed: 01/11/2023]
Abstract
Steroid hormones are synthesized in response to signaling cascades initiated by the trophic peptide hormones derived from the anterior pituitary. The mechanisms by which these peptide hormones regulate steroid hormone production are multifaceted and include controlling the transcription of steroidogenic genes, regulating cholesterol (substrate) uptake and transport, modulating steroidogenic enzyme activity, and controlling electron availability. Cytoskeletal polymers such as microfilaments and microtubules have also been implicated in regulating steroidogenesis. Of note, steroidogenesis is a multi-step process that occurs in two organelles, the endoplasmic reticulum (ER) and the mitochondrion. However, the precise mechanism by which substrates are delivered back and forth between these two organelles is unknown. In this review we will discuss the role of components of the cytoskeleton in conferring optimal steroidogenic potential. Finally, we present data that identifying a novel mechanism by which sphingosine-1-phosphate induces mitochondrial trafficking to promote steroidogenesis.
Collapse
Affiliation(s)
- Marion B. Sewer
- School of Biology and Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332-0230, USA e-mail:
| | - Donghui Li
- School of Biology and Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332-0230, USA e-mail:
| |
Collapse
|
37
|
Klei LR, Barchowsky A. Positive signaling interactions between arsenic and ethanol for angiogenic gene induction in human microvascular endothelial cells. Toxicol Sci 2008; 102:319-27. [PMID: 18182400 DOI: 10.1093/toxsci/kfn003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Arsenic in the drinking water may promote vascular diseases in millions of people worldwide through unresolved mechanisms. In addition, little is known of the effects of coexposures to arsenic and other common vasculature toxicants, such as alcohol. To investigate signaling interactions between arsenic and alcohols, primary human microvascular endothelial (HMVEC) cells were exposed to noncytotoxic concentrations of arsenite (1-5 microM) in the presence or absence of 0.1% ethanol (EtOH). Coexposure, but not exposure to either agent alone, rapidly increased active Fyn tyrosine kinase, tyrosine phosphorylation of a 109-kDa protein and serine phosphorylation of protein kinase C (PKC)delta. The 109-kDa protein was identified as PYK2, a regulator of vascular integrin signaling and an upstream activator of PKCdelta. Membrane localization of phospholipase Cgamma1 was increased by coexposure within 15 min, but not by either agent alone. In contrast, both agents equally increased membrane localization of Rac1-GTPase. Coexposure, but not exposure to either agent alone, induced transcript levels for the angiogenic genes, vascular endothelial cell growth factor (Vegfa) and insulin-like growth factor-1 (Igf1). However, EtOH inhibited arsenic-induced, nuclear factor-kappaB-driven interleukin-8 and collagen-1 expression. Differential effects of selective PKC inhibitors on induced gene expression combined with a lack of interaction for induction of hemeoxygenase-1 further demonstrated that arsenic-responsive signaling pathways differ in sensitivity to EtOH interactions. Finally, coexposure enhanced endothelial tube formation in in vitro angiogenesis assays. These data indicate that complex interactions occur between arsenic and EtOH exposures that functionally affect endothelial signaling for gene induction and remodeling stimuli.
Collapse
Affiliation(s)
- Linda R Klei
- Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania 15219, USA
| | | |
Collapse
|
38
|
Abstract
Steroid hormones are essential regulators of a vast number of physiological processes. The biosynthesis of these chemical messengers occurs in specialized steroidogenic tissues via a multi-step process that is catalyzed by members of the cytochrome P450 superfamily of monooxygenases and hydroxysteroid dehydrogenases. Though numerous signaling mediators, including cytokines and growth factors control steroidogenesis, trophic peptide hormones are the primary regulators of steroid hormone production. These peptide hormones activate a cAMP/cAMP-dependent kinase (PKA) signaling pathway, however, studies have shown that crosstalk between multiple signal transduction pathways and signaling molecules modulates optimal steroidogenic capacity. Sphingolipids such as ceramide, sphingosine, sphingosine-1-phosphate, sphingomyelin, and gangliosides have been shown to control the steroid hormone biosynthetic pathway at multiple levels, including regulating steroidogenic gene expression and activity as well as acting as second messengers in signaling cascades. In this review, we provide an overview of recent studies that have investigated the role of sphingolipids in adrenal, gonadal, and neural steroidogenesis.
Collapse
Affiliation(s)
- Natasha C Lucki
- School of Biology and Parker H, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 310 Ferst Drive, Atlanta, GA 30332-0230, USA
| | | |
Collapse
|