51
|
Hindle SJ, Bainton RJ. Barrier mechanisms in the Drosophila blood-brain barrier. Front Neurosci 2014; 8:414. [PMID: 25565944 PMCID: PMC4267209 DOI: 10.3389/fnins.2014.00414] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 11/24/2014] [Indexed: 12/12/2022] Open
Abstract
The invertebrate blood-brain barrier (BBB) field is growing at a rapid pace and, in recent years, studies have shown a physiologic and molecular complexity that has begun to rival its vertebrate counterpart. Novel mechanisms of paracellular barrier maintenance through G-protein coupled receptor signaling were the first demonstrations of the complex adaptive mechanisms of barrier physiology. Building upon this work, the integrity of the invertebrate BBB has recently been shown to require coordinated function of all layers of the compound barrier structure, analogous to signaling between the layers of the vertebrate neurovascular unit. These findings strengthen the notion that many BBB mechanisms are conserved between vertebrates and invertebrates, and suggest that novel findings in invertebrate model organisms will have a significant impact on the understanding of vertebrate BBB functions. In this vein, important roles in coordinating localized and systemic signaling to dictate organism development and growth are beginning to show how the BBB can govern whole animal physiologies. This includes novel functions of BBB gap junctions in orchestrating synchronized neuroblast proliferation, and of BBB secreted antagonists of insulin receptor signaling. These advancements and others are pushing the field forward in exciting new directions. In this review, we provide a synopsis of invertebrate BBB anatomy and physiology, with a focus on insights from the past 5 years, and highlight important areas for future study.
Collapse
Affiliation(s)
- Samantha J Hindle
- Department of Anesthesia and Perioperative Care, University of California, San Francisco San Francisco, CA, USA
| | - Roland J Bainton
- Department of Anesthesia and Perioperative Care, University of California, San Francisco San Francisco, CA, USA
| |
Collapse
|
52
|
Aikawa S, Hashimoto T, Kano K, Aoki J. Lysophosphatidic acid as a lipid mediator with multiple biological actions. J Biochem 2014; 157:81-9. [PMID: 25500504 DOI: 10.1093/jb/mvu077] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lysophosphatidic acid (LPA) is one of the simplest glycerophospholipids with one fatty acid chain and a phosphate group as a polar head. Although LPA had been viewed just as a metabolic intermediate in de novo lipid synthetic pathways, it has recently been paid much attention as a lipid mediator. LPA exerts many kinds of cellular processes, such as cell proliferation and smooth muscle contraction, through cognate G protein-coupled receptors. Because lipids are not coded by the genome directly, it is difficult to know their patho- and physiological roles. However, recent studies have identified several key factors mediating the biological roles of LPA, such as receptors and producing enzymes. In addition, studies of transgenic and gene knockout animals for these LPA-related genes, have revealed the biological significance of LPA. In this review we will summarize recent advances in the studies of LPA production and its roles in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Shizu Aikawa
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-Ku, Sendai 980-8578, Japan and CREST, Japan Science and Technology Corporation, 4-1-8, Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Takafumi Hashimoto
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-Ku, Sendai 980-8578, Japan and CREST, Japan Science and Technology Corporation, 4-1-8, Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Kuniyuki Kano
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-Ku, Sendai 980-8578, Japan and CREST, Japan Science and Technology Corporation, 4-1-8, Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-Ku, Sendai 980-8578, Japan and CREST, Japan Science and Technology Corporation, 4-1-8, Honcho, Kawaguchi, Saitama 332-0012, Japan Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-Ku, Sendai 980-8578, Japan and CREST, Japan Science and Technology Corporation, 4-1-8, Honcho, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
53
|
Morris AJ, Smyth SS. Lipid phosphate phosphatases: more than one way to put the brakes on LPA signaling? J Lipid Res 2014; 55:2195-7. [PMID: 25271297 DOI: 10.1194/jlr.c054957] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Andrew J Morris
- Division of Cardiovascular Medicine, University of Kentucky College of Medicine, Lexington Veterans Affairs Medical Center, Lexington, KY
| | - Susan S Smyth
- Division of Cardiovascular Medicine, University of Kentucky College of Medicine, Lexington Veterans Affairs Medical Center, Lexington, KY
| |
Collapse
|
54
|
Rivera-Pérez JA, Hadjantonakis AK. The Dynamics of Morphogenesis in the Early Mouse Embryo. Cold Spring Harb Perspect Biol 2014; 7:cshperspect.a015867. [PMID: 24968703 DOI: 10.1101/cshperspect.a015867] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
SUMMARYOver the past two decades, our understanding of mouse development from implantation to gastrulation has grown exponentially with an upsurge of genetic, molecular, cellular, and morphogenetic information. New discoveries have exalted the role of extraembryonic tissues in orchestrating embryonic patterning and axial specification. At the same time, the identification of unexpected morphogenetic processes occurring during mouse gastrulation has challenged established dogmas and brought new insights into the mechanisms driving germ layer formation. In this article, we summarize the key findings that have reinvigorated the contemporary view of early postimplantation mammalian development.
Collapse
Affiliation(s)
- Jaime A Rivera-Pérez
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan-Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
55
|
Bódis J, Papp S, Vermes I, Sulyok E, Tamás P, Farkas B, Zámbó K, Hatzipetros I, Kovács GL. "Platelet-associated regulatory system (PARS)" with particular reference to female reproduction. J Ovarian Res 2014; 7:55. [PMID: 24883111 PMCID: PMC4039651 DOI: 10.1186/1757-2215-7-55] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 04/21/2014] [Indexed: 02/06/2023] Open
Abstract
Background Blood platelets play an essential role in hemostasis, thrombosis and coagulation of blood. Beyond these classic functions their involvement in inflammatory, neoplastic and immune processes was also investigated. It is well known, that platelets have an armament of soluble molecules, factors, mediators, chemokines, cytokines and neurotransmitters in their granules, and have multiple adhesion molecules and receptors on their surface. Methods Selected relevant literature and own views and experiences as clinical observations have been used. Results Considering that platelets are indispensable in numerous homeostatic endocrine functions, it is reasonable to suppose that a platelet-associated regulatory system (PARS) may exist; internal or external triggers and/or stimuli may complement and connect regulatory pathways aimed towards target tissues and/or cells. The signal (PAF, or other tissue/cell specific factors) comes from the stimulated (by the e.g., hypophyseal hormones, bacteria, external factors, etc.) organs or cells, and activates platelets. Platelet activation means their aggregation, sludge formation, furthermore the release of the for-mentioned biologically very powerful factors, which can locally amplify and deepen the tissue specific cell reactions. If this process is impaired or inhibited for any reason, the specifically stimulated organ shows hypofunction. When PARS is upregulated, organ hyperfunction may occur that culminate in severe diseases. Conclusion Based on clinical and experimental evidences we propose that platelets modulate the function of hypothalamo-hypophyseal-ovarian system. Specifically, hypothalamic GnRH releases FSH from the anterior pituitary, which induces and stimulates follicular and oocyte maturation and steroid hormone secretion in the ovary. At the same time follicular cells enhance PAF production. Through these pathways activated platelets are accumulated in the follicular vessels surrounding the follicle and due to its released soluble molecules (factors, mediators, chemokines, cytokines, neurotransmitters) locally increase oocyte maturation and hormone secretion. Therefore we suggest that platelets are not only a small participant but may be the conductor or active mediator of this complex regulatory system which has several unrevealed mechanisms. In other words platelets are corpuscular messengers, or are more than a member of the family providing hemostasis.
Collapse
Affiliation(s)
- József Bódis
- Department of Obstetrics and Gynecology, University of Pécs, 7624 Pécs Édesanyák útja 17, Hungary ; HAS-UP Human reproduction scientific research group, 7624 Pécs Édesanyák útja 17, Hungary
| | - Szilárd Papp
- Department of Obstetrics and Gynecology, University of Pécs, 7624 Pécs Édesanyák útja 17, Hungary
| | - István Vermes
- Institiute of Diagnostics, Faculty of Health Sciences, University of Pécs, 7400 Kaposvár, Szent Imre u. 14/b, Hungary
| | - Endre Sulyok
- Faculty of Health Sciences, University of Pécs, 7621 Pécs Vörösmarty u. 4, Hungary
| | - Péter Tamás
- Department of Obstetrics and Gynecology, University of Pécs, 7624 Pécs Édesanyák útja 17, Hungary
| | - Bálint Farkas
- Department of Obstetrics and Gynecology, University of Pécs, 7624 Pécs Édesanyák útja 17, Hungary
| | - Katalin Zámbó
- Department of Nuclear Medicine, University of Pécs, 7624 Pécs Ifjúság u. 13, Hungary
| | - Ioannis Hatzipetros
- Department of Obstetrics and Gynecology, University of Pécs, 7624 Pécs Édesanyák útja 17, Hungary
| | - Gábor L Kovács
- Department of Laboratory Medicine, University of Pécs, 7624 Pécs Ifjúság u. 13, Hungary ; Szentagothai Research Centre, University of Pécs, 7624 Pécs, Ifjúsag u. 20., Hungary
| |
Collapse
|
56
|
Motiejūnaitė R, Aranda J, Kazlauskas A. Pericytes prevent regression of endothelial cell tubes by accelerating metabolism of lysophosphatidic acid. Microvasc Res 2014; 93:62-71. [DOI: 10.1016/j.mvr.2014.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 01/16/2014] [Accepted: 03/19/2014] [Indexed: 01/10/2023]
|
57
|
Chemical and genetic tools to explore S1P biology. Curr Top Microbiol Immunol 2014; 378:55-83. [PMID: 24728593 PMCID: PMC7120161 DOI: 10.1007/978-3-319-05879-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The zwitterionic lysophospholipid Sphingosine 1-Phosphate (S1P) is a pleiotropic mediator of physiology and pathology. The synthesis, transport, and degradation of S1P are tightly regulated to ensure that S1P is present in the proper concentrations in the proper location. The binding of S1P to five G protein-coupled S1P receptors regulates many physiological systems, particularly the immune and vascular systems. Our understanding of the functions of S1P has been aided by the tractability of the system to both chemical and genetic manipulation. Chemical modulators have been generated to affect most of the known components of S1P biology, including agonists of S1P receptors and inhibitors of enzymes regulating S1P production and degradation. Genetic knockouts and manipulations have been similarly engineered to disrupt the functions of individual S1P receptors or enzymes involved in S1P metabolism. This chapter will focus on the development and utilization of these chemical and genetic tools to explore the complex biology surrounding S1P and its receptors, with particular attention paid to the in vivo findings that these tools have allowed for.
Collapse
|
58
|
Abstract
Sphingosine 1-phosphate (S1P) is a lipid mediator formed by the metabolism of sphingomyelin. In vertebrates, S1P is secreted into the extracellular environment and signals via G protein-coupled S1P receptors to regulate cell-cell and cell-matrix adhesion, and thereby influence cell migration, differentiation and survival. The expression and localization of S1P receptors is dynamically regulated and controls vascular development, vessel stability and immune cell trafficking. In addition, crucial events during embryogenesis, such as angiogenesis, cardiogenesis, limb development and neurogenesis, are regulated by S1P signalling. Here, and in the accompanying poster, we provide an overview of S1P signalling in development and in disease.
Collapse
Affiliation(s)
- Karen Mendelson
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | | | | |
Collapse
|
59
|
Panchatcharam M, Salous AK, Brandon J, Miriyala S, Wheeler J, Patil P, Sunkara M, Morris AJ, Escalante-Alcalde D, Smyth SS. Mice with targeted inactivation of ppap2b in endothelial and hematopoietic cells display enhanced vascular inflammation and permeability. Arterioscler Thromb Vasc Biol 2014; 34:837-45. [PMID: 24504738 DOI: 10.1161/atvbaha.113.302335] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Lipid phosphate phosphatase 3 (LPP3), encoded by the PPAP2B gene, is an integral membrane enzyme that dephosphorylates, and thereby terminates, the G-protein-coupled receptor-mediated signaling actions of lysophosphatidic acid (LPA) and sphingosine-1-phosphate. LPP3 is essential for normal vascular development in mice, and a common PPAP2B polymorphism is associated with increased risk of coronary artery disease in humans. Herein, we investigate the function of endothelial LPP3 to understand its role in the development and human disease. APPROACH AND RESULTS We developed mouse models with selective LPP3 deficiency in endothelial and hematopoietic cells. Tyrosine kinase Tek promoter-mediated inactivation of Ppap2b resulted in embryonic lethality because of vascular defects. LPP3 deficiency in adult mice, achieved using a tamoxifen-inducible Cre transgene under the control of the Tyrosine kinase Tek promoter, enhanced local and systemic inflammatory responses. Endothelial, but not hematopoietic, cell LPP3 deficiency led to significant increases in vascular permeability at baseline and enhanced sensitivity to inflammation-induced vascular leak. Endothelial barrier function was restored by pharmacological or genetic inhibition of either LPA production by the circulating lysophospholipase D autotaxin or of G-protein-coupled receptor-dependent LPA signaling. CONCLUSIONS Our results identify a role for the autotaxin/LPA-signaling nexus as a mediator of endothelial permeability in inflammation and demonstrate that LPP3 limits these effects. These findings have implications for therapeutic targets to maintain vascular barrier function in inflammatory states.
Collapse
Affiliation(s)
- Manikandan Panchatcharam
- From the Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, KY (M.P., A.K.S., J.B., S.M., J.W., P.P., M.S., A.J.M., S.S.S.); División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México DF, Mexico (E.-A.); and Medical Service, Lexington VA Medical Center, Lexington, KY (A.J.M., S.S.S.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Smyth SS, Mueller P, Yang F, Brandon JA, Morris AJ. Arguing the case for the autotaxin-lysophosphatidic acid-lipid phosphate phosphatase 3-signaling nexus in the development and complications of atherosclerosis. Arterioscler Thromb Vasc Biol 2014; 34:479-86. [PMID: 24482375 DOI: 10.1161/atvbaha.113.302737] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The structurally simple glycero- and sphingo-phospholipids, lysophosphatidic acid (LPA) and sphingosine-1-phosphate, serve as important receptor-active mediators that influence blood and vascular cell function and are positioned to influence the events that contribute to the progression and complications of atherosclerosis. Growing evidence from preclinical animal models has implicated LPA, LPA receptors, and key enzymes involved in LPA metabolism in pathophysiologic events that may underlie atherosclerotic vascular disease. These observations are supported by genetic analysis in humans implicating a lipid phosphate phosphatase as a novel risk factor for coronary artery disease. In this review, we summarize current understanding of LPA production, metabolism, and signaling as may be relevant for atherosclerotic and other vascular disease.
Collapse
Affiliation(s)
- Susan S Smyth
- From the Veterans Affairs Medical Center, Cardiovascular Medicine Service, Lexington, KY (S.S.S., A.J.M.); and Division of Cardiovascular Medicine, Gill Heart Institute, University of Kentucky, Lexington, KY (S.S.S., P.M., F.Y., J.A.B., A.J.M.)
| | | | | | | | | |
Collapse
|
61
|
Gutiérrez-Martínez E, Fernández-Ulibarri I, Lázaro-Diéguez F, Johannes L, Pyne S, Sarri E, Egea G. Lipid phosphate phosphatase 3 participates in transport carrier formation and protein trafficking in the early secretory pathway. J Cell Sci 2013; 126:2641-55. [PMID: 23591818 DOI: 10.1242/jcs.117705] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The inhibition of phosphatidic acid phosphatase (PAP) activity by propanolol indicates that diacylglycerol (DAG) is required for the formation of transport carriers at the Golgi and for retrograde trafficking to the ER. Here we report that the PAP2 family member lipid phosphate phosphatase 3 (LPP3, also known as PAP2b) localizes in compartments of the secretory pathway from ER export sites to the Golgi complex. The depletion of human LPP3: (i) reduces the number of tubules generated from the ER-Golgi intermediate compartment and the Golgi, with those formed from the Golgi being longer in LPP3-silenced cells than in control cells; (ii) impairs the Rab6-dependent retrograde transport of Shiga toxin subunit B from the Golgi to the ER, but not the anterograde transport of VSV-G or ssDsRed; and (iii) induces a high accumulation of Golgi-associated membrane buds. LPP3 depletion also reduces levels of de novo synthesized DAG and the Golgi-associated DAG contents. Remarkably, overexpression of a catalytically inactive form of LPP3 mimics the effects of LPP3 knockdown on Rab6-dependent retrograde transport. We conclude that LPP3 participates in the formation of retrograde transport carriers at the ER-Golgi interface, where it transitorily cycles, and during its route to the plasma membrane.
Collapse
Affiliation(s)
- Enric Gutiérrez-Martínez
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
62
|
Bianchini L, Birtwisle L, Saâda E, Bazin A, Long E, Roussel JF, Michiels JF, Forest F, Dani C, Myklebost O, Birtwisle-Peyrottes I, Pedeutour F. Identification of PPAP2B as a novel recurrent translocation partner gene of HMGA2 in lipomas. Genes Chromosomes Cancer 2013; 52:580-90. [PMID: 23508853 DOI: 10.1002/gcc.22055] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 02/06/2013] [Accepted: 02/06/2013] [Indexed: 12/19/2022] Open
Abstract
Most lipomas are characterized by translocations involving the HMGA2 gene in 12q14.3. These rearrangements lead to the fusion of HMGA2 with an ectopic sequence from the translocation chromosome partner. Only five fusion partners of HMGA2 have been identified in lipomas so far. The identification of novel fusion partners of HMGA2 is important not only for diagnosis in soft tissue tumors but also because these genes might have an oncogenic role in other tumors. We observed that t(1;12)(p32;q14) was the second most frequent translocation in our series of lipomas after t(3;12)(q28;q14.3). We detected overexpression of HMGA2 mRNA and protein in all t(1;12)(p32;q14) lipomas. We used a fluorescence in situ hybridization-based positional cloning strategy to characterize the 1p32 breakpoint. In 11 cases, we identified PPAP2B, a member of the lipid phosphate phosphatases family as the 1p32 target gene. Reverse transcription-polymerase chain reaction analysis followed by nucleotide sequencing of the fusion transcript indicated that HMGA2 3' untranslated region (3'UTR) fused with exon 6 of PPAP2B in one case. In other t(1;12) cases, the breakpoint was extragenic, located in the 3'region flanking PPAP2B 3'UTR. Moreover, in one case showing a t(1;6)(p32;p21) we observed a rearrangement of PPAP2B and HMGA1, which suggests that HMGA1 might also be a fusion partner for PPAP2B. Our results also revealed that adipocytic differentiation of human mesenchymal stem cells derived from adipose tissue was associated with a significant decrease in PPAP2B mRNA expression suggesting that PPAP2B might play a role in adipogenesis.
Collapse
Affiliation(s)
- Laurence Bianchini
- Laboratory of Solid Tumors Genetics, Nice University Hospital, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284/INSERM U1081, University of Nice-Sophia Antipolis, Nice, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Morishige J, Uto Y, Hori H, Satouchi K, Yoshiomoto T, Tokumura A. Lysophosphatidic acid produced by hen egg white lysophospholipase D induces vascular development on extraembryonic membranes. Lipids 2013; 48:251-62. [PMID: 23381130 DOI: 10.1007/s11745-013-3765-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 01/09/2013] [Indexed: 01/23/2023]
Abstract
Lysophosphatidic acid (lysoPtdOH), a lysophospholipid mediator, exerts diverse physiological effects, including angiogenesis, through its specific G-protein-coupled receptors. Previously, we showed that unfertilized hen egg white contains polyunsaturated fatty acid-rich lysoPtdOH and lysophospholipase D (lysoPLD). Here, we examined whether lysoPtdOH was produced by lysoPLD in the presence and absence of a hen fertilized ovum and what the physiological role of lysoPtdOH in hen egg white is. Mass spectrometry showed that fertilized hen egg white contained about 8 μM lysoPtdOH before incubation with an ovum, mainly comprised of 18:1- (12.6 %), 18:2- (37.8 %) and 20:4-molecular species (41.5 %). In an early gestation period, the lysoPtdOH was increased up to 9.6 μM, concomitant with a decrease in the level of polyunsaturated lysophosphatidylcholine (lysoPtdCho). Moreover, lysoPtdOH-degrading activities were found in egg white and the vitelline membrane, showing that these enzymes control lysoPtdOH levels in egg white. In an egg yolk angiogenesis assay, two lysoPtdOH receptor antagonists, Ki16425 and N-palmitoyl serine phosphoric acid (NASP), inhibited blood vessel formation induced by exogenously added 18:1-lysoPtdOH and its precursor lysoPtdCho on the hen yolk sac. Ki16425 and NASP also inhibited blood vessel formation in the chorioallantoic membrane (CAM). Furthermore, the relatively higher levels of LPA₁, LPA₂, LPA₄ and LPA₆ mRNA were present in the yolk sac and CAM. These results suggest that lysoPtdOH produced from lysoPtdCho by the action of lysoPLD in hen egg white is involved in the formation of blood vessel networks through several lysoPtdOH receptors on various extraembryonic membranes, including the yolk sac membrane and CAM.
Collapse
Affiliation(s)
- Junichi Morishige
- Department of Pharmaceutical Health Chemistry, Institute of Health Biosciences, University of Tokushima Graduate School, 1-78-1 Shomachi, Tokushima 770-8505, Japan
| | | | | | | | | | | |
Collapse
|
64
|
Xu H, Yang Y, Tang X, Zhao M, Liang F, Xu P, Hou B, Xing Y, Bao X, Fan X. Bergmann glia function in granule cell migration during cerebellum development. Mol Neurobiol 2013; 47:833-44. [PMID: 23329344 DOI: 10.1007/s12035-013-8405-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 01/07/2013] [Indexed: 12/17/2022]
Abstract
Granule cell migration influences the laminar structure of the cerebellum and thereby affects cerebellum function. Bergmann glia are derived from radial glial cells and aid in granule cell radial migration by providing a scaffold for migration and by mediating interactions between Bergmann glia and granule cells. In this review, we summarize Bergmann glia characteristics and the mechanisms underlying the effect of Bergmann glia on the radial migration of granule neurons in the cerebellum. Furthermore, we will focus our discussion on the important factors involved in glia-mediated radial migration so that we may elucidate the possible mechanistic pathways used by Bergmann glia to influence granule cell migration during cerebellum development.
Collapse
Affiliation(s)
- Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University, Chongqing 400038, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Xu H, Yang Y, Tang X, Zhao M, Liang F, Xu P, Hou B, Xing Y, Bao X, Fan X. Bergmann glia function in granule cell migration during cerebellum development. Mol Neurobiol 2013. [PMID: 23329344 DOI: 10.1007/s12035‐013‐8405‐y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Granule cell migration influences the laminar structure of the cerebellum and thereby affects cerebellum function. Bergmann glia are derived from radial glial cells and aid in granule cell radial migration by providing a scaffold for migration and by mediating interactions between Bergmann glia and granule cells. In this review, we summarize Bergmann glia characteristics and the mechanisms underlying the effect of Bergmann glia on the radial migration of granule neurons in the cerebellum. Furthermore, we will focus our discussion on the important factors involved in glia-mediated radial migration so that we may elucidate the possible mechanistic pathways used by Bergmann glia to influence granule cell migration during cerebellum development.
Collapse
Affiliation(s)
- Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University, Chongqing 400038, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Ren H, Panchatcharam M, Mueller P, Escalante-Alcalde D, Morris AJ, Smyth SS. Lipid phosphate phosphatase (LPP3) and vascular development. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1831:126-32. [PMID: 22835522 PMCID: PMC3683602 DOI: 10.1016/j.bbalip.2012.07.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 07/15/2012] [Accepted: 07/16/2012] [Indexed: 01/12/2023]
Abstract
Lipid phosphate phosphatases (LPP) are integral membrane proteins with broad substrate specificity that dephosphorylate lipid substrates including phosphatidic acid, lysophosphatidic acid, ceramide 1-phosphate, sphingosine 1-phosphate, and diacylglycerol pyrophosphate. Although the three mammalian enzymes (LPP1-3) demonstrate overlapping catalytic activities and substrate preferences in vitro, the phenotypes of mice with targeted inactivation of the Ppap2 genes encoding the LPP enzymes reveal nonredundant functions. A specific role for LPP3 in vascular development has emerged from studies of mice lacking Ppap2b. A meta-analysis of multiple, large genome-wide association studies identified a single nucleotide polymorphism in PPAP2B as a novel predictor of coronary artery disease. In this review, we will discuss the evidence that links LPP3 to vascular development and disease and evaluate potential molecular mechanisms. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
Affiliation(s)
- H Ren
- The Gill Heart Institute, Division of Cardiovascular Medicine, Lexington, KY 40536-0200, USA
| | | | | | | | | | | |
Collapse
|
67
|
Ile KE, Renault AD. Compartmentalizing the embryo: lipids and septate junction mediated barrier function. Fly (Austin) 2012; 7:18-22. [PMID: 23221483 PMCID: PMC3660746 DOI: 10.4161/fly.22938] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Lipid phosphate phosphatases (LPPs) are a class of enzymes that can dephosphorylate a number of lysophopholipids in vitro. Analysis of knockouts of LPP family members has demonstrated striking but diverse developmental roles for these enzymes. LPP3 is required for mouse vascular development while the Drosophila LPPs Wunen (Wun) and Wunen2 (Wun2) are required during embryogenesis for germ cell migration and survival. In a recent publication we examined if these fly LPPs have further developmental roles and found that Wun is required for proper tracheal formation. In particular we highlight a role for Wun in septate junction mediated barrier function in the tracheal system. In this paper we discuss further the possible mechanisms by which LPPs may influence barrier activity.
Collapse
Affiliation(s)
- Kristina E Ile
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | | |
Collapse
|
68
|
Gaaya A, Poirier O, Mougenot N, Hery T, Atassi F, Marchand A, Saulnier-Blache JS, Amour J, Vogt J, Lompré AM, Soubrier F, Nadaud S. Plasticity-related gene-1 inhibits lysophosphatidic acid-induced vascular smooth muscle cell migration and proliferation and prevents neointima formation. Am J Physiol Cell Physiol 2012; 303:C1104-14. [DOI: 10.1152/ajpcell.00051.2012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Plasticity-related gene-1 (PRG-1) protects neuronal cells from lysophosphatidic acid (LPA) effects. In vascular smooth muscle cells (VSMCs), LPA was shown to induce phenotypic modulation in vitro and vascular remodeling in vivo. Thus we explored the role of PRG-1 in modulating VSMC response to LPA. PCR, Western blot, and immunofluorescence experiments showed that PRG-1 is expressed in rat and human vascular media. PRG-1 expression was strongly inhibited in proliferating compared with quiescent VSMCs both in vitro and in vivo (medial vs. neointimal VSMCs), suggesting that PRG-1 expression is dependent on the cell phenotype. In vitro, adenovirus-mediated overexpression of PRG-1 specifically inhibited LPA-induced rat VSMC proliferation and migration but not platelet-derived growth factor-induced proliferation. This effect was abolished by mutation of a conserved histidine in the lipid phosphate phosphatase family that is essential for interaction with lipid phosphates. In vivo, balloon-induced neointimal formation in rat carotid was significantly decreased in vessels infected with PRG-1 adenovirus compared with β-galactosidase adenovirus (−71%; P < 0.05). PRG-1 overexpression abolished the activation of the p42/p44 signaling pathway in LPA-stimulated rat VSMCs in culture and in balloon-injured rat carotids. Taken together, these findings provide the first evidence of a protective role of PRG-1 in the vascular media under pathophysiological conditions.
Collapse
Affiliation(s)
- Amira Gaaya
- INSERM, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche UMR_S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
- UPMC Univ Paris 06, Université Pierre et Marie Curie, UMR-S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
| | - Odette Poirier
- INSERM, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche UMR_S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
- UPMC Univ Paris 06, Université Pierre et Marie Curie, UMR-S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
| | - Nathalie Mougenot
- INSERM, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche UMR_S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
- INSERM, Faculté de Médecine Pitié-Salpétrière, PECMV-IFR14, Paris, France
| | - Tiphaine Hery
- INSERM, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche UMR_S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
- UPMC Univ Paris 06, Université Pierre et Marie Curie, UMR-S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
| | - Fabrice Atassi
- INSERM, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche UMR_S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
- UPMC Univ Paris 06, Université Pierre et Marie Curie, UMR-S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
| | - Alexandre Marchand
- INSERM, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche UMR_S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
- UPMC Univ Paris 06, Université Pierre et Marie Curie, UMR-S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
| | - Jean-Sébastien Saulnier-Blache
- INSERM, U1048/I2MC, Toulouse, France
- Université Toulouse III Paul Sabatier, Institut de Médecine Moléculaire de Rangueil, Toulouse, France
| | - Julien Amour
- INSERM, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche UMR_S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
- Department of Anesthesiology and Critical Care Medicine, Centre Hospitalier Universitaire Pitié-Salpêtrière, Paris, France; and
| | - Johannes Vogt
- Institute for Microanatomy and Neurobiology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Anne-Marie Lompré
- INSERM, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche UMR_S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
- UPMC Univ Paris 06, Université Pierre et Marie Curie, UMR-S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
| | - Florent Soubrier
- INSERM, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche UMR_S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
- UPMC Univ Paris 06, Université Pierre et Marie Curie, UMR-S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
| | - Sophie Nadaud
- INSERM, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche UMR_S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
- UPMC Univ Paris 06, Université Pierre et Marie Curie, UMR-S 956, Faculté de Médecine Pitié-Salpétrière, Paris, France
| |
Collapse
|
69
|
Panchatcharam M, Miriyala S, Salous A, Wheeler J, Dong A, Mueller P, Sunkara M, Escalante-Alcalde D, Morris AJ, Smyth SS. Lipid phosphate phosphatase 3 negatively regulates smooth muscle cell phenotypic modulation to limit intimal hyperplasia. Arterioscler Thromb Vasc Biol 2012; 33:52-9. [PMID: 23104851 DOI: 10.1161/atvbaha.112.300527] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE The lipid phosphate phosphatase 3 (LPP3) degrades bioactive lysophospholipids, including lysophosphatidic acid and sphingosine-1-phosphate, and thereby terminates their signaling effects. Although emerging evidence links lysophosphatidic acid to atherosclerosis and vascular injury responses, little is known about the role of vascular LPP3. The goal of this study was to determine the role of LPP3 in the development of vascular neointima formation and smooth muscle cells (SMC) responses. METHODS AND RESULTS We report that LPP3 is expressed in vascular SMC after experimental arterial injury. Using gain- and loss-of-function approaches, we establish that a major function of LPP3 in isolated SMC cells is to attenuate proliferation (extracellular signal-regulated kinases) activity, Rho activation, and migration in response to serum and lysophosphatidic acid. These effects are at least partially a consequence of LPP3-catalyzed lysophosphatidic acid hydrolysis. Mice with selective inactivation of LPP3 in SMC display an exaggerated neointimal response to injury. CONCLUSIONS Our observations suggest that LPP3 serves as an intrinsic negative regulator of SMC phenotypic modulation and inflammation after vascular injury, in part, by regulating lysophospholipid signaling. These findings may provide a mechanistic link to explain the association between a PPAP2B polymorphism and coronary artery disease risk.
Collapse
Affiliation(s)
- Manikandan Panchatcharam
- Division of Cardiovascular Medicine, The Gill Heart Institute, 255 BBRSB, 741 S. Limestone St, Lexington, KY 40536-0200, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Sánchez-Sánchez R, Morales-Lázaro SL, Baizabal JM, Sunkara M, Morris AJ, Escalante-Alcalde D. Lack of lipid phosphate phosphatase-3 in embryonic stem cells compromises neuronal differentiation and neurite outgrowth. Dev Dyn 2012; 241:953-64. [PMID: 22434721 DOI: 10.1002/dvdy.23779] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Bioactive lipids such as lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) have been recently described as important regulators of pluripotency and differentiation of embryonic stem (ES) cells and neural progenitors. Due to the early lethality of LPP3, an enzyme that regulates the levels and biological activities of the aforementioned lipids, it has been difficult to assess its participation in early neural differentiation and neuritogenesis. RESULTS We find that Ppap2b(-/-) (Lpp3(-/-) ) ES cells differentiated in vitro into spinal neurons show a considerable reduction in the amount of neural precursors and young neurons formed. In addition, differentiated Lpp3(-/-) neurons exhibit impaired neurite outgrowth. Surprisingly, when Lpp3(-/-) ES cells were differentiated, an unexpected appearance of smooth muscle actin-positive cells was observed, an event that was partially dependent upon phosphorylated sphingosines. CONCLUSIONS Our data show that LPP3 plays a fundamental role during spinal neuron differentiation from ES and that it also participates in regulating neurite and axon outgrowth.
Collapse
Affiliation(s)
- Roberto Sánchez-Sánchez
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México
| | | | | | | | | | | |
Collapse
|
71
|
Abstract
The allantois is the embryonic precursor of the umbilical cord in mammals and is one of several embryonic regions, including the yolk sac and dorsal aorta, that undergoes vasculogenesis, the de novo formation of blood vessels. Despite its importance in establishing the chorioallantoic placenta and umbilical circulation, the allantois frequently is overlooked in embryologic studies. Nonetheless, recent studies demonstrate that vasculogenesis, vascular remodeling, and angiogenesis are essential allantois functions in the establishment of the chorioallantoic placenta. Here, we review blood vessel formation in the murine allantois, highlighting the expression of genes and involvement of pathways common to vasculogenesis or angiogenesis in other parts of the embryo. We discuss experimental techniques available for manipulation of the allantois that are unavailable for yolk sac or dorsal aorta, and review how this system has been used as a model system to discover new genes and mechanisms involved in vessel formation. Finally, we discuss the potential of the allantois as a model system to provide insights into disease and therapeutics.
Collapse
|
72
|
Kok BPC, Venkatraman G, Capatos D, Brindley DN. Unlike two peas in a pod: lipid phosphate phosphatases and phosphatidate phosphatases. Chem Rev 2012; 112:5121-46. [PMID: 22742522 DOI: 10.1021/cr200433m] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Bernard P C Kok
- Signal Transduction Research Group, Department of Biochemistry, School of Translational Medicine, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | | | | | | |
Collapse
|
73
|
Aaltonen N, Lehtonen M, Varonen K, Goterris GA, Laitinen JT. Lipid phosphate phosphatase inhibitors locally amplify lysophosphatidic acid LPA1 receptor signalling in rat brain cryosections without affecting global LPA degradation. BMC Pharmacol 2012; 12:7. [PMID: 22686545 PMCID: PMC3418163 DOI: 10.1186/1471-2210-12-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 06/11/2012] [Indexed: 11/10/2022] Open
Abstract
Background Lysophosphatidic acid (LPA) is a signalling phospholipid with multiple biological functions, mainly mediated through specific G protein-coupled receptors. Aberrant LPA signalling is being increasingly implicated in the pathology of common human diseases, such as arteriosclerosis and cancer. The lifetime of the signalling pool of LPA is controlled by the equilibrium between synthesizing and degradative enzymatic activity. In the current study, we have characterized these enzymatic pathways in rat brain by pharmacologically manipulating the enzymatic machinery required for LPA degradation. Results In rat brain cryosections, the lifetime of bioactive LPA was found to be controlled by Mg2+-independent, N-ethylmaleimide-insensitive phosphatase activity, attributed to lipid phosphate phosphatases (LPPs). Pharmacological inhibition of this LPP activity amplified LPA1 receptor signalling, as revealed using functional autoradiography. Although two LPP inhibitors, sodium orthovanadate and propranolol, locally amplified receptor responses, they did not affect global brain LPA phosphatase activity (also attributed to Mg2+-independent, N-ethylmaleimide-insensitive phosphatases), as confirmed by Pi determination and by LC/MS/MS. Interestingly, the phosphate analog, aluminium fluoride (AlFx-) not only irreversibly inhibited LPP activity thereby potentiating LPA1 receptor responses, but also totally prevented LPA degradation, however this latter effect was not essential in order to observe AlFx--dependent potentiation of receptor signalling. Conclusions We conclude that vanadate- and propranolol-sensitive LPP activity locally guards the signalling pool of LPA whereas the majority of brain LPA phosphatase activity is attributed to LPP-like enzymatic activity which, like LPP activity, is sensitive to AlFx- but resistant to the LPP inhibitors, vanadate and propranolol.
Collapse
Affiliation(s)
- Niina Aaltonen
- School of Pharmacy, University of Eastern Finland, P,O, Box 1627, 70211, Kuopio, Finland.
| | | | | | | | | |
Collapse
|
74
|
Ile KE, Tripathy R, Goldfinger V, Renault AD. Wunen, a Drosophila lipid phosphate phosphatase, is required for septate junction-mediated barrier function. Development 2012; 139:2535-46. [PMID: 22675212 DOI: 10.1242/dev.077289] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Lipid phosphate phosphatases (LPPs) are integral membrane enzymes that regulate the levels of bioactive lipids such as sphingosine 1-phosphate and lysophosphatidic acid. The Drosophila LPPs Wunen (Wun) and Wunen-2 (Wun2) have a well-established role in regulating the survival and migration of germ cells. We now show that wun has an essential tissue-autonomous role in development of the trachea: the catalytic activity of Wun is required to maintain septate junction (SJ) paracellular barrier function, loss of which causes failure to accumulate crucial luminal components, suggesting a role for phospholipids in SJ function. We find that the integrity of the blood-brain barrier is also lost in wun mutants, indicating that loss of SJ function is not restricted to the tracheal system. Furthermore, by comparing the rescue ability of different LPP homologs we show that wun function in the trachea is distinct from its role in germ cell migration.
Collapse
Affiliation(s)
- Kristina E Ile
- Max Planck Institute for Developmental Biology, Spemannstr. 35, 72076 Tübingen, Germany
| | | | | | | |
Collapse
|
75
|
Watson ED, Hughes M, Simmons DG, Natale DR, Sutherland AE, Cross JC. Cell-cell adhesion defects in Mrj mutant trophoblast cells are associated with failure to pattern the chorion during early placental development. Dev Dyn 2011; 240:2505-19. [DOI: 10.1002/dvdy.22755] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2011] [Indexed: 11/12/2022] Open
|
76
|
A novel perspective on stem cell homing and mobilization: review on bioactive lipids as potent chemoattractants and cationic peptides as underappreciated modulators of responsiveness to SDF-1 gradients. Leukemia 2011; 26:63-72. [PMID: 21886175 DOI: 10.1038/leu.2011.242] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hematopoietic stem progenitor cells (HSPCs) respond robustly to α-chemokine stromal-derived factor-1 (SDF-1) gradients, and blockage of CXCR4, a seven-transmembrane-spanning G(αI)-protein-coupled SDF-1 receptor, mobilizes HSPCs into peripheral blood. Although the SDF-1-CXCR4 axis has an unquestionably important role in the retention of HSPCs in bone marrow (BM), new evidence shows that, in addition to SDF-1, the migration of HSPCs is directed by gradients of the bioactive lipids sphingosine-1 phosphate and ceramide-1 phosphate. Furthermore, the SDF-1 gradient may be positively primed/modulated by cationic peptides (C3a anaphylatoxin and cathelicidin) and, as previously demonstrated, HSPCs respond robustly even to very low SDF-1 gradients in the presence of priming factors. In this review, we discuss the role of bioactive lipids in stem cell trafficking and the consequences of HSPC priming by cationic peptides. Together, these phenomena support a picture in which the SDF-1-CXCR4 axis modulates homing, BM retention and mobilization of HSPCs in a more complex way than previously envisioned.
Collapse
|
77
|
Mir-290-295 deficiency in mice results in partially penetrant embryonic lethality and germ cell defects. Proc Natl Acad Sci U S A 2011; 108:14163-8. [PMID: 21844366 DOI: 10.1073/pnas.1111241108] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mir-290 through mir-295 (mir-290-295) is a mammalian-specific microRNA (miRNA) cluster that, in mice, is expressed specifically in early embryos and embryonic germ cells. Here, we show that mir-290-295 plays important roles in embryonic development as indicated by the partially penetrant lethality of mutant embryos. In addition, we show that in surviving mir-290-295-deficient embryos, female but not male fertility is compromised. This impairment in fertility arises from a defect in migrating primordial germ cells and occurs equally in male and female mutant animals. Male mir-290-295(-/-) mice, due to the extended proliferative lifespan of their germ cells, are able to recover from this initial germ cell loss and are fertile. Female mir-290-295(-/-) mice are unable to recover and are sterile, due to premature ovarian failure.
Collapse
|
78
|
Kleger A, Liebau S, Lin Q, von Wichert G, Seufferlein T. The impact of bioactive lipids on cardiovascular development. Stem Cells Int 2011; 2011:916180. [PMID: 21876704 PMCID: PMC3159013 DOI: 10.4061/2011/916180] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 06/05/2011] [Indexed: 12/30/2022] Open
Abstract
Lysophospholipids comprise a group of bioactive molecules with multiple biological functions. The cardinal members of this signalling molecule group are sphingosylphosphorylcholine (SPC), lysophosphatidic acid (LPA), and sphingosine 1-phosphate (S1P) which are, at least in part, homologous to each other. Bioactive lipids usually act via G-protein coupled receptors (GPCRs), but can also function as direct intracellular messengers. Recently, it became evident that bioactive lipids play a role during cellular differentiation development. SPC induces mesodermal differentiation of mouse ES cells and differentiation of promyelocytic leukemia cells, by a mechanism being critically dependent on MEK-ERK signalling. LPA stimulates the clonal expansion of neurospheres from neural stem/progenitor cells and induces c-fos via activation of mitogen- and stress-activated protein kinase 1 (MSK1) in ES cells. S1P acts on hematopoietic progenitor cells as a chemotactic factor and has also been found to be critical for cardiac and skeletal muscle regeneration. Furthermore, S1P promotes cardiogenesis and similarly activates Erk signalling in mouse ES cells. Interestingly, S1P may also act to maintain human stem cell pluripotency. Both LPA and S1P positively regulate the proliferative capacity of murine ES cells. In this paper we will focus on the differential and developmental impact of lysophospholipids on cardiovascular development.
Collapse
Affiliation(s)
- Alexander Kleger
- Department of Internal Medicine I, University of Ulm, 89081 Ulm, Germany
| | | | | | | | | |
Collapse
|
79
|
Bréart B, Ramos-Perez WD, Mendoza A, Salous AK, Gobert M, Huang Y, Adams RH, Lafaille JJ, Escalante-Alcalde D, Morris AJ, Schwab SR. Lipid phosphate phosphatase 3 enables efficient thymic egress. ACTA ACUST UNITED AC 2011; 208:1267-78. [PMID: 21576386 PMCID: PMC3173249 DOI: 10.1084/jem.20102551] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Lipid phosphate phosphatase 3 in endothelial and epithelial cells promotes efficient T cell emigration from the thymus to the periphery. The signaling lipid sphingosine-1-phosphate (S1P) stabilizes the vasculature, directs lymphocyte egress from lymphoid organs, and shapes inflammatory responses. However, little is known about how S1P distribution is controlled in vivo, and it is not clear how a ubiquitously made lipid functions as a signal that requires precise spatial and temporal control. We have found that lipid phosphate phosphatase 3 (LPP3) enables efficient export of mature T cells from the thymus into circulation, and several lines of evidence suggest that LPP3 promotes exit by destroying thymic S1P. Although five additional S1P-degrading enzymes are expressed in the thymus, they cannot compensate for the loss of LPP3. Moreover, conditional deletion of LPP3 in either epithelial cells or endothelial cells is sufficient to inhibit egress. These results suggest that S1P generation and destruction are tightly regulated and that LPP3 is essential to establish the balance.
Collapse
Affiliation(s)
- Béatrice Bréart
- Program in Molecular Pathogenesis and Department of Pathology, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Chatterjee I, Humtsoe JO, Kohler EE, Sorio C, Wary KK. Lipid phosphate phosphatase-3 regulates tumor growth via β-catenin and CYCLIN-D1 signaling. Mol Cancer 2011; 10:51. [PMID: 21569306 PMCID: PMC3112429 DOI: 10.1186/1476-4598-10-51] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 05/11/2011] [Indexed: 12/13/2022] Open
Abstract
Background The acquisition of proliferative and invasive phenotypes is considered a hallmark of neoplastic transformation; however, the underlying mechanisms are less well known. Lipid phosphate phosphatase-3 (LPP3) not only catalyzes the dephosphorylation of the bioactive lipid sphingosine-1-phosphate (S1P) to generate sphingosine but also may regulate embryonic development and angiogenesis via the Wnt pathway. The goal of this study was to determine the role of LPP3 in tumor cells. Results We observed increased expression of LPP3 in glioblastoma primary tumors and in U87 and U118 glioblastoma cell lines. We demonstrate that LPP3-knockdown inhibited both U87 and U118 glioblastoma cell proliferation in culture and tumor growth in xenograft assays. Biochemical experiments provided evidence that LPP3-knockdown reduced β-catenin, CYCLIN-D1, and CD133 expression, with a concomitant increase in phosphorylated β-catenin. In a converse experiment, the forced expression of LPP3 in human colon tumor (SW480) cells potentiated tumor growth via increased β-catenin stability and CYCLIN-D1 synthesis. In contrast, elevated expression of LPP3 had no tumorigenic effects on primary cells. Conclusions These results demonstrate for the first time an unexpected role of LPP3 in regulating glioblastoma progression by amplifying β-catenin and CYCLIN-D1 activities.
Collapse
Affiliation(s)
- Ishita Chatterjee
- Dept of Pharmacology, University of Illinois, 835 S Wolcott, Room E403, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
81
|
López-Juárez A, Morales-Lázaro S, Sánchez-Sánchez R, Sunkara M, Lomelí H, Velasco I, Morris AJ, Escalante-Alcalde D. Expression of LPP3 in Bergmann glia is required for proper cerebellar sphingosine-1-phosphate metabolism/signaling and development. Glia 2011; 59:577-89. [PMID: 21319224 PMCID: PMC3196773 DOI: 10.1002/glia.21126] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Accepted: 11/22/2010] [Indexed: 02/02/2023]
Abstract
Bioactive lipids serve as intracellular and extracellular mediators in cell signaling in normal and pathological conditions. Here we describe that an important regulator of some of these lipids, the lipid phosphate phosphatase-3 (LPP3), is abundantly expressed in specific plasma membrane domains of Bergmann glia (BG), a specialized type of astrocyte with key roles in cerebellum development and physiology. Mice selectively lacking expression of LPP3/Ppap2b in the nervous system are viable and fertile but exhibit defects in postnatal cerebellum development and modifications in the cytoarchitecture and arrangement of BG with a mild non-progressive motor coordination defect. Lipid and gene profiling studies in combination with pharmacological treatments suggest that most of these effects are associated with alterations in sphingosine-1-phosphate (S1P) metabolism and signaling. Altogether our data indicate that LPP3 participates in several aspects of neuron-glia communication required for proper cerebellum development.
Collapse
Affiliation(s)
- Alejandro López-Juárez
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México
| | - Sara Morales-Lázaro
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México
| | - Roberto Sánchez-Sánchez
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México
| | | | - Hilda Lomelí
- Instituto de Biotecnología, Universidad Nacional Autónoma de México
| | - Iván Velasco
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México
| | | | - Diana Escalante-Alcalde
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México
| |
Collapse
|
82
|
In vitro and in vivo comparison of viral and cellular internal ribosome entry sites for bicistronic vector expression. Gene Ther 2011; 18:631-6. [PMID: 21368899 DOI: 10.1038/gt.2011.11] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bicistronic vectors are essential to achieve efficient expression of multiple genes in gene therapy protocols and biomedical applications. Internal ribosome entry site (IRES) elements have been utilized to initiate expression of an additional protein from a bicistronic vector. The IRES element commonly used in current bicistronic vectors originates from the encephalomyocarditis virus (EMCV). As IRES-mediated translation is dependent on availability of IRES trans-acting factors, which vary between cell types and species, adequate gene expression from the EMCV IRES element is not always achieved. To identify a novel IRES element that mediates gene expression consistently with a higher efficiency than the EMCV IRES, we tested 13 bicistronic reporter constructs containing different viral and cellular IRES elements. The in vitro screening in human and mouse fibroblast and hepatocarcinoma cells revealed that the vascular endothelial growth factor and type 1 collagen-inducible protein (VCIP) IRES was the only IRES element that directed translation more efficiently than the EMCV IRES in all cell lines. Furthermore, the VCIP IRES initiated greater reporter expression levels than the EMCV IRES in transfected mouse livers. These results suggest that VCIP-IRES containing vectors improve gene expression compared with those harboring an EMCV-IRES. This could increase the potential benefits of bicistronic vectors for experimental and therapeutic purposes.
Collapse
|
83
|
Regulation of phosphatidic Acid metabolism by sphingolipids in the central nervous system. J Lipids 2010; 2011:342576. [PMID: 21490799 PMCID: PMC3068476 DOI: 10.1155/2011/342576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 10/14/2010] [Indexed: 12/12/2022] Open
Abstract
This paper explores the way ceramide, sphingosine, ceramide 1-phosphate, and sphingosine 1-phosphate modulate the generation of second lipid messengers from phosphatidic acid in two experimental models of the central nervous system: in vertebrate rod outer segments prepared from dark-adapted retinas as well as in rod outer segments prepared from light-adapted retinas and in rat cerebral cortex synaptosomes under physiological aging conditions. Particular attention is paid to lipid phosphate phosphatase, diacylglycerol lipase, and monoacylglycerol lipase. Based on the findings reported in this paper, it can be concluded that proteins related to phototransduction phenomena are involved in the effects derived from sphingosine 1-phosphate/sphingosine or ceramide 1-phosphate/ceramide and that age-related changes occur in the metabolism of phosphatidic acid from cerebral cortex synaptosomes in the presence of either sphingosine 1-phosphate/sphingosine or ceramide 1-phosphate/ceramide. The present paper demonstrates, in two different models of central nervous system, how sphingolipids influence phosphatidic acid metabolism under different physiological conditions such as light and aging.
Collapse
|
84
|
Liu N, Enkemann SA, Liang P, Hersmus R, Zanazzi C, Huang J, Wu C, Chen Z, Looijenga LHJ, Keefe DL, Liu L. Genome-wide gene expression profiling reveals aberrant MAPK and Wnt signaling pathways associated with early parthenogenesis. J Mol Cell Biol 2010; 2:333-44. [PMID: 20926514 DOI: 10.1093/jmcb/mjq029] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mammalian parthenogenesis could not survive but aborted during mid-gestation, presumably because of lack of paternal gene expression. To understand the molecular mechanisms underlying the failure of parthenogenesis at early stages of development, we performed global gene expression profiling and functional analysis of parthenogenetic blastocysts in comparison with those of blastocysts from normally fertilized embryos. Parthenogenetic blastocysts exhibited changes in the expression of 749 genes, of which 214 had lower expression and 535 showed higher expressions than fertilized embryos using a minimal 1.8-fold change as a cutoff. Genes important for placenta development were decreased in their expression in parthenote blastocysts. Some maternally expressed genes were up-regulated and paternal-related genes were down-regulated. Moreover, aberrantly increased Wnt signaling and reduced mitogen-activated protein kinase (MAPK) signaling were associated with early parthenogenesis. The protein level of extracellular signal-regulated kinase 2 (ERK2) was low in parthenogenetic blastocysts compared with that of fertilized blastocysts 120 h after fertilization. 6-Bromoindirubin-3'-oxime, a specific glycogen synthase kinase-3 (GSK-3) inhibitor, significantly decreased embryo hatching. The expression of several imprinted genes was altered in parthenote blastocysts. Gene expression also linked reduced expression of Xist to activation of X chromosome. Our findings suggest that failed X inactivation, aberrant imprinting, decreased ERK/MAPK signaling and possibly elevated Wnt signaling, and reduced expression of genes for placental development collectively may contribute to abnormal placenta formation and failed fetal development in parthenogenetic embryos.
Collapse
Affiliation(s)
- Na Liu
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Zhou L, Picard D, Ra YS, Li M, Northcott PA, Hu Y, Stearns D, Hawkins C, Taylor MD, Rutka J, Der SD, Huang A. Silencing of thrombospondin-1 is critical for myc-induced metastatic phenotypes in medulloblastoma. Cancer Res 2010; 70:8199-210. [PMID: 20876797 DOI: 10.1158/0008-5472.can-09-4562] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mechanisms by which c-Myc (Myc) amplification confers aggressive medulloblastoma phenotypes are poorly defined. Here, we show using orthotopic models that high Myc expression promotes cell migration/invasion and induces metastatic tumors, which recapitulate aggressive histologic features of Myc-amplified primary human medulloblastoma. Using ChIP-chip analysis, we identified cell migration and adhesion genes, including Tsp-1/THBS1, ING4, PVRL3, and PPAP2B, as Myc-bound loci in medulloblastoma cells. Expression of Tsp-1 was most consistently and robustly diminished in medulloblastoma cell lines and primary human tumors with high Myc expression (n = 101, P = 0.032). Strikingly, stable Tsp-1 expression significantly attenuated in vitro transformation and invasive/migratory properties of high Myc-expressing medulloblastoma cells without altering cell proliferation, whereas RNA interference-mediated Myc knockdown was consistently accompanied by increased Tsp-1 levels and reduced cell migration and invasion in medulloblastoma cells. Chromatin immunoprecipitation (ChIP) assays revealed colocalization of Myc and obligate partner Max and correlated diminished RNA polymerase II occupancy (∼3-fold decrease, P < 0.01) with increased Myc binding at a core Tsp-1 promoter. Reporter gene and/or gel shift assays confirmed direct repression of Tsp-1 transcription by Myc and also identified JPO2, a Myc interactor associated with metastatic medulloblastoma, as a cofactor in Myc-mediated Tsp-1 repression. These findings indicate the Myc-regulatory network targets Tsp-1 via multiple mechanisms in medulloblastoma transformation, and highlight a novel critical role for Tsp-1 in Myc-mediated aggressive medulloblastoma phenotypes.
Collapse
Affiliation(s)
- Limei Zhou
- Sonia and Arthur Labatt Brain Tumor Research Centre, Hospital for Sick Children, University Avenue, Toronto, Ontario, Canada M5G 1X8
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Abstract
Lysophosphatidic acid (LPA) is a potent lipid mediator with a wide variety of biological actions mediated through G protein-coupled receptors (LPA(1-6)). LPA(4) has been identified as a G(13) protein-coupled receptor, but its physiological role is unknown. Here we show that a subset of LPA(4)-deficient embryos did not survive gestation and displayed hemorrhages and/or edema in many organs at multiple embryonic stages. The blood vessels of bleeding LPA(4)-deficient embryos were often dilated. The recruitment of mural cells, namely smooth muscle cells and pericytes, was impaired. Consistently, Matrigel plug assays showed decreased mural cell coverage of endothelial cells in the neovessels of LPA(4)-deficient adult mice. In situ hybridization detected Lpa4 mRNA in the endothelium of some vasculatures. Similarly, the lymphatic vessels of edematous embryos were dilated. These results suggest that LPA(4) regulates establishment of the structure and function of blood and lymphatic vessels during mouse embryogenesis. Considering the critical role of autotaxin (an enzyme involved in LPA production) and Gα(13) in vascular development, we suggest that LPA(4) provides a link between these 2 molecules.
Collapse
|
87
|
Miura K, Miura S, Yamasaki K, Shimada T, Kinoshita A, Niikawa N, Yoshiura KI, Masuzaki H. The possibility of microarray-based analysis using cell-free placental mRNA in maternal plasma. Prenat Diagn 2010; 30:849-61. [DOI: 10.1002/pd.2570] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
88
|
Abstract
Lysophosphatidic acid (LPA; 1- or 2-acyl-sn-glycerol-3-phosphate) is a phospholipid that is involved in numerous normal physiological and pathological processes such as brain development, blood vessel formation, embryo implantation, hair growth, neuropathic pain, lung fibrosis and colon cancer. Most of these functions are mediated by G protein-coupled receptors (GPCRs) specific to LPA. So far, six GPCRs for LPA have been identified: LPA(1)/Edg2, LPA(2)/Edg4, LPA(3)/Edg7, LPA(4)/GPR23/P2Y9, LPA(5)/GPR92 and LPA(6)/P2Y5. An intracellular target of LPA has also been proposed. Among the LPA receptors, LPA(3) is unique in that it is activated significantly by a specific form of LPA (2-acyl LPA with unsaturated fatty acids) and is expressed in a limited number of tissues such as the reproductive organs. Recent studies have shown that LPA(3)-mediated LPA signaling is essential for proper embryo implantation and have revealed an unexpected genetic linkage between LPA and prostaglandin signaling. Here we review recent advances in the study of LPA(3), especially studies using LPA(3)-deficient mice. In addition, we focus on the agonists and antagonists that are specific to each LPA receptor as important tools for the functional study of LPA signaling.
Collapse
Affiliation(s)
- Kotaro Hama
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, Japan
| | | |
Collapse
|
89
|
Lipid phosphate phosphatase 3 stabilization of beta-catenin induces endothelial cell migration and formation of branching point structures. Mol Cell Biol 2010; 30:1593-606. [PMID: 20123964 DOI: 10.1128/mcb.00038-09] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Endothelial cell (EC) migration, cell-cell adhesion, and the formation of branching point structures are considered hallmarks of angiogenesis; however, the underlying mechanisms of these processes are not well understood. Lipid phosphate phosphatase 3 (LPP3) is a recently described p120-catenin-associated integrin ligand localized in adherens junctions (AJs) of ECs. Here, we tested the hypothesis that LPP3 stimulates beta-catenin/lymphoid enhancer binding factor 1 (beta-catenin/LEF-1) to induce EC migration and formation of branching point structures. In subconfluent ECs, LPP3 induced expression of fibronectin via beta-catenin/LEF-1 signaling in a phosphatase and tensin homologue (PTEN)-dependent manner. In confluent ECs, depletion of p120-catenin restored LPP3-mediated beta-catenin/LEF-1 signaling. Depletion of LPP3 resulted in destabilization of beta-catenin, which in turn reduced fibronectin synthesis and deposition, which resulted in inhibition of EC migration. Accordingly, reexpression of beta-catenin but not p120-catenin in LPP3-depleted ECs restored de novo synthesis of fibronectin, which mediated EC migration and formation of branching point structures. In confluent ECs, however, a fraction of p120-catenin associated and colocalized with LPP3 at the plasma membrane, via the C-terminal cytoplasmic domain, thereby limiting the ability of LPP3 to stimulate beta-catenin/LEF-1 signaling. Thus, our study identified a key role for LPP3 in orchestrating PTEN-mediated beta-catenin/LEF-1 signaling in EC migration, cell-cell adhesion, and formation of branching point structures.
Collapse
|
90
|
Morris AJ, Selim S, Salous A, Smyth SS. Blood relatives: dynamic regulation of bioactive lysophosphatidic acid and sphingosine-1-phosphate metabolism in the circulation. Trends Cardiovasc Med 2009; 19:135-40. [PMID: 19818950 DOI: 10.1016/j.tcm.2009.07.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Lysophosphatidic acid and sphingosine 1-phosphate are bioactive lipid mediators with potent effects on cardiovascular development and vascular function. New studies define dynamic mechanisms that maintain physiologically relevant levels of both lipids in the blood. We review the mechanisms controlling the production, metabolism, and distribution of these lipids between vascular cells, circulating blood components, and the plasma.
Collapse
Affiliation(s)
- Andrew J Morris
- The Gill Heart Institute, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| | | | | | | |
Collapse
|
91
|
Giusto NM, Pasquaré SJ, Salvador GA, Ilincheta de Boschero MG. Lipid second messengers and related enzymes in vertebrate rod outer segments. J Lipid Res 2009; 51:685-700. [PMID: 19828910 DOI: 10.1194/jlr.r001891] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Rod outer segments (ROSs) are specialized light-sensitive organelles in vertebrate photoreceptor cells. Lipids in ROS are of considerable importance, not only in providing an adequate environment for efficient phototransduction, but also in originating the second messengers involved in signal transduction. ROSs have the ability to adapt the sensitivity and speed of their responses to ever-changing conditions of ambient illumination. A major contributor to this adaptation is the light-driven translocation of key signaling proteins into and out of ROS. The present review shows how generation of the second lipid messengers from phosphatidylcholine, phosphatidic acid, and diacylglycerol is modulated by the different illumination states in the vertebrate retina. Findings suggest that the light-induced translocation of phototransduction proteins influences the enzymatic activities of phospholipase D, lipid phosphate phosphatase, diacylglyceride lipase, and diacylglyceride kinase, all of which are responsible for the generation of the second messenger molecules.
Collapse
Affiliation(s)
- Norma M Giusto
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Bahía Blanca, Argentina.
| | | | | | | |
Collapse
|
92
|
Regulation of myoblast differentiation by the nuclear envelope protein NET39. Mol Cell Biol 2009; 29:5800-12. [PMID: 19704009 DOI: 10.1128/mcb.00684-09] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recently, several transmembrane proteins of the nuclear envelope have been implicated in regulation of signaling and gene expression. Here we demonstrate that the nuclear lamina-associated nuclear envelope transmembrane protein NET39 (Ppapdc3) functions as a negative regulator of myoblast differentiation, in part through effects on mTOR signaling. We found that NET39 is highly expressed in cardiac and skeletal muscle tissues and becomes strongly upregulated during cultured myoblast differentiation. Knockdown of NET39 by RNA interference in myoblasts strongly promoted differentiation, whereas overexpression of NET39 repressed myogenesis. Proteomic analysis of NET39 complexes immunoprecipitated from myotubes, in combination with other methods, identified mTOR as an interaction partner of NET39. We found that ectopic expression of NET39 in myoblasts negatively regulated myogenesis by diminishing mTOR activity, which in turn decreased insulin-like growth factor II production and autocrine signaling. Our results indicate that NET39 is part of the regulatory machinery for myogenesis and raise the possibility that it may be important for muscle homeostasis.
Collapse
|
93
|
Morris AJ, Panchatcharam M, Cheng HY, Federico L, Fulkerson Z, Selim S, Miriyala S, Escalante-Alcalde D, Smyth SS. Regulation of blood and vascular cell function by bioactive lysophospholipids. J Thromb Haemost 2009; 7 Suppl 1:38-43. [PMID: 19630765 PMCID: PMC2801156 DOI: 10.1111/j.1538-7836.2009.03405.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lysophosphatidic acid (LPA), its sphingolipid homolog sphingosine 1-phosphate (S1P) and several other related molecules constitute a family of bioactive lipid phosphoric acids that function as receptor-active mediators with roles in cell growth, differentiation, inflammation, immunomodulation, apoptosis and development. LPA and S1P are present in physiologically relevant concentrations in the circulation. In isolated cell culture systems or animal models, these lipids exert a range of effects that suggest that S1P and LPA could play important roles in maintaining normal vascular homeostasis and in vascular injury responses. LPA and S1P act on a series of G protein-coupled receptors, and LPA may also be an endogenous regulator of PPARgamma activity. In this review, we discuss potential roles for lysolipid signaling in the vasculature and mechanisms by which these bioactive lipids could contribute to cardiovascular disease.
Collapse
Affiliation(s)
- A J Morris
- The Gill Heart Institute, Division of Cardiovascular Medicine, Lexington, KY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Lipid phosphate phosphohydrolase type 1 (LPP1) degrades extracellular lysophosphatidic acid in vivo. Biochem J 2009; 419:611-8. [PMID: 19215222 DOI: 10.1042/bj20081888] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
LPA (lysophosphatidic acid) is a lipid mediator that stimulates cell proliferation and growth, and is involved in physiological and pathological processes such as wound healing, platelet activation, angiogenesis and the growth of tumours. Therefore defining the mechanisms of LPA production and degradation are of interest in understanding the regulation of these processes. Extracellular LPA synthesis is relatively well understood, whereas the mechanisms of its degradation are not. One route of LPA degradation is dephosphorylation. A candidate enzyme is the integral membrane exophosphatase LPP1 (lipid phosphate phosphohydrolase type 1). In the present paper, we report the development of a mouse wherein the LPP1 gene (Ppap2a) was disrupted. The homozygous mice, which are phenotypically unremarkable, generally lack Ppap2a mRNA, and multiple tissues exhibit a substantial (35-95%) reduction in LPA phosphatase activity. Compared with wild-type littermates, Ppap2a(tr/tr) animals have increased levels of plasma LPA, and LPA injected intravenously is metabolized at a 4-fold lower rate. Our results demonstrate that LPA is rapidly metabolized in the bloodstream and that LPP1 is an important determinant of this turnover. These results indicate that LPP1 is a catabolic enzyme for LPA in vivo.
Collapse
|
95
|
Boles MK, Wilkinson BM, Maxwell A, Lai L, Mills AA, Nishijima I, Salinger AP, Moskowitz I, Hirschi KK, Liu B, Bradley A, Justice MJ. A mouse chromosome 4 balancer ENU-mutagenesis screen isolates eleven lethal lines. BMC Genet 2009; 10:12. [PMID: 19267930 PMCID: PMC2670824 DOI: 10.1186/1471-2156-10-12] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 03/06/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND ENU-mutagenesis is a powerful technique to identify genes regulating mammalian development. To functionally annotate the distal region of mouse chromosome 4, we performed an ENU-mutagenesis screen using a balancer chromosome targeted to this region of the genome. RESULTS We isolated 11 lethal lines that map to the region of chromosome 4 between D4Mit117 and D4Mit281. These lines form 10 complementation groups. The majority of lines die during embryonic development between E5.5 and E12.5 and display defects in gastrulation, cardiac development, and craniofacial development. One line displayed postnatal lethality and neurological defects, including ataxia and seizures. CONCLUSION These eleven mutants allow us to query gene function within the distal region of mouse chromosome 4 and demonstrate that new mouse models of mammalian developmental defects can easily and quickly be generated and mapped with the use of ENU-mutagenesis in combination with balancer chromosomes. The low number of mutations isolated in this screen compared with other balancer chromosome screens indicates that the functions of genes in different regions of the genome vary widely.
Collapse
Affiliation(s)
- Melissa K Boles
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Shockley KR, Lazarenko OP, Czernik PJ, Rosen CJ, Churchill GA, Lecka-Czernik B. PPARgamma2 nuclear receptor controls multiple regulatory pathways of osteoblast differentiation from marrow mesenchymal stem cells. J Cell Biochem 2009; 106:232-46. [PMID: 19115254 DOI: 10.1002/jcb.21994] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rosiglitazone (Rosi), a member of the thiazolidinedione class of drugs used to treat type 2 diabetes, activates the adipocyte-specific transcription factor peroxisome proliferator-activated receptor gamma (PPARgamma). This activation causes bone loss in animals and humans, at least in part due to suppression of osteoblast differentiation from marrow mesenchymal stem cells (MSC). In order to identify mechanisms by which PPARgamma2 suppresses osteoblastogenesis and promotes adipogenesis in MSC, we have analyzed the PPARgamma2 transcriptome in response to Rosi. A total of 4,252 transcriptional changes resulted when Rosi (1 microM) was applied to the U-33 marrow stromal cell line stably transfected with PPARgamma2 (U-33/gamma2) as compared to non-induced U-33/gamma2 cells. Differences between U-33/gamma2 and U-33 cells stably transfected with empty vector (U-33/c) comprised 7,928 transcriptional changes, independent of Rosi. Cell type-, time- and treatment-specific gene clustering uncovered distinct patterns of PPARgamma2 transcriptional control of MSC lineage commitment. The earliest changes accompanying Rosi activation of PPARgamma2 included effects on Wnt, TGFbeta/BMP and G-protein signaling activities, as well as sustained induction of adipocyte-specific gene expression and lipid metabolism. While suppression of osteoblast phenotype is initiated by a diminished expression of osteoblast-specific signaling pathways, induction of the adipocyte phenotype is initiated by adipocyte-specific transcriptional regulators. This indicates that distinct mechanisms govern the repression of osteogenesis and the stimulation of adipogenesis. The co-expression patterns found here indicate that PPARgamma2 has a dominant role in controlling osteoblast differentiation and suggests numerous gene-gene interactions that could lead to the identification of a "master" regulatory scheme directing this process.
Collapse
Affiliation(s)
- Keith R Shockley
- The Jackson Laboratory, 600 Main Street, Bar Harbor, Maine 04609, USA
| | | | | | | | | | | |
Collapse
|
97
|
Downs KM, Inman KE, Jin DX, Enders AC. The Allantoic Core Domain: new insights into development of the murine allantois and its relation to the primitive streak. Dev Dyn 2009; 238:532-53. [PMID: 19191225 PMCID: PMC2966891 DOI: 10.1002/dvdy.21862] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The whereabouts and properties of the posterior end of the primitive streak have not been identified in any species. In the mouse, the streak's posterior terminus is assumed to be confined to the embryonic compartment, and to give rise to the allantois, which links the embryo to its mother during pregnancy. In this study, we have refined our understanding of the biology of the murine posterior primitive streak and its relation to the allantois. Through a combination of immunostaining and morphology, we demonstrate that the primitive streak spans the posterior extraembryonic and embryonic regions at the onset of the neural plate stage ( approximately 7.0 days postcoitum, dpc). Several hours later, the allantoic bud emerges from the extraembryonic component of the primitive streak (XPS). Then, possibly in collaboration with overlying allantois-associated extraembryonic visceral endoderm, the XPS establishes a germinal center within the allantois, named here the Allantoic Core Domain (ACD). Microsurgical removal of the ACD beyond headfold (HF) stages resulted in the formation of allantoic regenerates that lacked the ACD and failed to elongate; nevertheless, vasculogenesis and vascular patterning proceeded. In situ and transplantation fate mapping demonstrated that, from HF stages onward, the ACD's progenitor pool contributed to the allantois exclusive of the proximal flanks. By contrast, the posterior intraembryonic primitive streak (IPS) provided the flanks. Grafting the ACD into T(C)/T(C) hosts, whose allantoises are significantly foreshortened, restored allantoic elongation. These results revealed that the ACD is essential for allantoic elongation, but the cues required for vascularization lie outside of it. On the basis of these and previous findings, we conclude that the posterior primitive streak of the mouse conceptus is far more complex than was previously believed. Our results provide new directives for addressing the origin and development of the umbilical cord, and establish a novel paradigm for investigating the fetal/placental relationship.
Collapse
Affiliation(s)
- Karen M Downs
- Department of Anatomy, University of Wisconsin-Madison School of Medicine and Public Health, 1300 University Avenue, Madison, WI 53706, USA.
| | | | | | | |
Collapse
|
98
|
Abstract
One of the major lipid biology discoveries in last decade was the broad range of physiological activities of lysophospholipids that have been attributed to the actions of lysophospholipid receptors. The most well characterized lysophospholipids are lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P). Documented cellular effects of these lipid mediators include growth-factor-like effects on cells, such as proliferation, survival, migration, adhesion, and differentiation. The mechanisms for these actions are attributed to a growing family of 7-transmembrane, G protein-coupled receptors (GPCRs). Their pathophysiological actions include immune modulation, neuropathic pain modulation, platelet aggregation, wound healing, vasopressor activity, and angiogenesis. Here we provide a brief introduction to receptor-mediated lysophospholipid signaling and physiology, and then discuss potential therapeutic roles in human diseases.
Collapse
|
99
|
Federico L, Pamuklar Z, Smyth SS, Morris AJ. Therapeutic potential of autotaxin/lysophospholipase d inhibitors. Curr Drug Targets 2008; 9:698-708. [PMID: 18691016 DOI: 10.2174/138945008785132439] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Lysophosphatidic acids (LPAs) are structurally simple lipid phosphate esters with a widely appreciated role as extracellular signaling molecules. LPA binds to selective cell surface receptors to promote cell growth, survival, motility and differentiation. Studies using LPA receptor knockout mice and experimental therapeutics targeting these receptors identify roles for LPA signaling in processes that include cardiovascular disease and function, angiogenesis, reproduction, cancer progression and neuropathic pain. These studies identify considerable functional redundancy between these receptors and raise the possibility that additional lysophosphatidic acid receptors remain to be identified. LPA is present in the blood and other biological fluids at physiologically relevant concentrations and can likely be rapidly generated and degraded in different locations, for example at sites of inflammation, vascular injury and thrombosis or in the tumor micro environment. Recent work identifies a secreted enzyme, autotaxin (ATX), as the key component of an extracellular pathway for generation of lysophosphatidic acid by lysophospholipase D catalyzed hydrolysis of lysophospholipid substrates. In contrast to the apparently redundant functions of LPA receptors, studies using ATX knock out and transgenic mice indicate that this enzyme is uniquely required for LPA signaling during early development and serves as the primary determinant of circulating LPA levels in adult animals. Accordingly, pharmacological inhibition of ATX may be a viable and potentially effective way to interfere with LPA signaling in the cardiovascular system and possibly other settings such as tumor metastasis for therapeutic benefit. In this review we provide an update on recent advances in defining roles for LPA signaling in major disease processes and discuss recent progress in understanding the regulation and function of autotaxin focusing on strategies for the identification and initial evaluation of small molecule autotaxin inhibitors.
Collapse
Affiliation(s)
- Lorenzo Federico
- Division of Cardiovascular Medicine, The Gill Heart Institute, 900 S. Limestone Street, 326 CTW Building, Lexington, KY 40536-0200, USA
| | | | | | | |
Collapse
|
100
|
Cohen TV, Klarmann KD, Sakchaisri K, Cooper JP, Kuhns D, Anver M, Johnson PF, Williams SC, Keller JR, Stewart CL. The lamin B receptor under transcriptional control of C/EBPepsilon is required for morphological but not functional maturation of neutrophils. Hum Mol Genet 2008; 17:2921-33. [PMID: 18621876 PMCID: PMC2536505 DOI: 10.1093/hmg/ddn191] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Accepted: 07/03/2008] [Indexed: 11/14/2022] Open
Abstract
The lamin B receptor (LBR) is an integral nuclear envelope protein that interacts with chromatin and has homology to sterol reductases. Mutations in LBR result in Pelger-Huët anomaly and HEM-Greenberg skeletal dysplasia, whereas in mice Lbr mutations result in ichthyosis. To further understand the function of the LBR and its role in disease, we derived a novel mouse model with a gene-trap insertion into the Lbr locus (Lbr(GT/GT)). Phenotypically, the Lbr(GT/GT) mice are similar to ichthyosis mice. The Lbr(GT/GT) granulocytes lack a mature segmented nucleus and have a block in late maturation. Despite these changes in nuclear morphology, the innate granulocyte immune function in the killing of Staphylococcus aureus bacteria appears to be intact. Granulocyte differentiation requires the transcription factor C/EBPepsilon. We identified C/EBPepsilon binding sites within the Lbr promoter and used EMSAs and luciferase assays to show that Lbr is transcriptionally regulated by C/EBPepsilon. Our findings indicate that the Lbr(GT/GT) mice are a model for Pelger-Huët anomaly and that Lbr, under transcriptional regulation of C/EBPepsilon, is necessary for morphological but not necessarily functional granulocyte maturation.
Collapse
Affiliation(s)
| | - Kimberly D. Klarmann
- Cancer and Developmental Biology Laboratory, CCR
- Basic Research Program, Laboratory of Cancer Prevention, SAIC-Frederick, Inc
| | | | - Jason P. Cooper
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | - Miriam Anver
- Laboratory Animal Sciences Program, Pathology/Histotechnology Laboratory, SAIC-Frederick, National Cancer Institute, Frederick, MD 21702, USA
| | | | - Simon C. Williams
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Jonathan R. Keller
- Cancer and Developmental Biology Laboratory, CCR
- Basic Research Program, Laboratory of Cancer Prevention, SAIC-Frederick, Inc
| | | |
Collapse
|