1
|
Chia ZJ, Kumarapperuma H, Zhang R, Little PJ, Kamato D. Smad transcription factors as mediators of 7 transmembrane G protein-coupled receptor signalling. Acta Pharmacol Sin 2024:10.1038/s41401-024-01413-6. [PMID: 39506064 DOI: 10.1038/s41401-024-01413-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
The Smad transcription factors are well known for their role at the core of transforming growth factor-β (TGF-β) signalling. However, recent evidence shows that the Smad transcription factors play a vital role downstream of other classes of receptors including G protein-coupled receptors (GPCR). The versatility of Smad transcription factors originated from the two regions that can be differently activated by the TGF-β receptor superfamily or through the recruitment of intracellular kinases stimulated by other receptors classes such as GPCRs. The classic GPCR signalling cascade is further expanded to conditional adoption of the Smad transcription factor under the stimulation of Akt, demonstrating the unique involvement of the Smad transcription factor in GPCR signalling pathways in disease environments. In this review, we provide a summary of the signalling pathways of the Smad transcription factors as an important downstream mediator of GPCRs, presenting exciting opportunities for discovering new therapeutic targets for diseases.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Hirushi Kumarapperuma
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
| | - Ruizhi Zhang
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia
- School of Environment and Science, Griffith Sciences, Griffith University, Nathan, QLD, Australia
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD, Australia.
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, Australia.
- School of Environment and Science, Griffith Sciences, Griffith University, Nathan, QLD, Australia.
| |
Collapse
|
2
|
Fischer C, Schreiber Y, Nitsch R, Vogt J, Thomas D, Geisslinger G, Tegeder I. Lysophosphatidic Acid Receptors LPAR5 and LPAR2 Inversely Control Hydroxychloroquine-Evoked Itch and Scratching in Mice. Int J Mol Sci 2024; 25:8177. [PMID: 39125747 PMCID: PMC11312285 DOI: 10.3390/ijms25158177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Lysophosphatidic acids (LPAs) evoke nociception and itch in mice and humans. In this study, we assessed the signaling paths. Hydroxychloroquine was injected intradermally to evoke itch in mice, which evoked an increase of LPAs in the skin and in the thalamus, suggesting that peripheral and central LPA receptors (LPARs) were involved in HCQ-evoked pruriception. To unravel the signaling paths, we assessed the localization of candidate genes and itching behavior in knockout models addressing LPAR5, LPAR2, autotaxin/ENPP2 and the lysophospholipid phosphatases, as well as the plasticity-related genes Prg1/LPPR4 and Prg2/LPPR3. LacZ reporter studies and RNAscope revealed LPAR5 in neurons of the dorsal root ganglia (DRGs) and in skin keratinocytes, LPAR2 in cortical and thalamic neurons, and Prg1 in neuronal structures of the dorsal horn, thalamus and SSC. HCQ-evoked scratching behavior was reduced in sensory neuron-specific Advillin-LPAR5-/- mice (peripheral) but increased in LPAR2-/- and Prg1-/- mice (central), and it was not affected by deficiency of glial autotaxin (GFAP-ENPP2-/-) or Prg2 (PRG2-/-). Heat and mechanical nociception were not affected by any of the genotypes. The behavior suggested that HCQ-mediated itch involves the activation of peripheral LPAR5, which was supported by reduced itch upon treatment with an LPAR5 antagonist and autotaxin inhibitor. Further, HCQ-evoked calcium fluxes were reduced in primary sensory neurons of Advillin-LPAR5-/- mice. The results suggest that LPA-mediated itch is primarily mediated via peripheral LPAR5, suggesting that a topical LPAR5 blocker might suppress "non-histaminergic" itch.
Collapse
Affiliation(s)
- Caroline Fischer
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.F.); (D.T.); (G.G.)
| | - Yannick Schreiber
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt am Main, Germany;
| | - Robert Nitsch
- Institute for Translational Neuroscience, Medical Faculty, WWU Münster, 48149 Münster, Germany;
| | - Johannes Vogt
- Department of Molecular and Translational Neurosciences, Institute for Anatomy and Center of Molecular Medicine Cologne (CMMC), and Cologne Excellence Cluster for Aging associated Diseases (CECAD), University of Cologne, 50923 Köln, Germany;
| | - Dominique Thomas
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.F.); (D.T.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt am Main, Germany;
| | - Gerd Geisslinger
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.F.); (D.T.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt am Main, Germany;
- Fraunhofer Cluster of Excellence of Immune Mediated Diseases (CIMD), 60596 Frankfurt am Main, Germany
| | - Irmgard Tegeder
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.F.); (D.T.); (G.G.)
| |
Collapse
|
3
|
Dacheux MA, Norman DD, Tigyi GJ, Lee SC. Emerging roles of lysophosphatidic acid receptor subtype 5 (LPAR5) in inflammatory diseases and cancer. Pharmacol Ther 2023; 245:108414. [PMID: 37061203 DOI: 10.1016/j.pharmthera.2023.108414] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/17/2023]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid mediator that regulates a variety of cellular functions such as cell proliferation, migration, survival, calcium mobilization, cytoskeletal rearrangements, and neurite retraction. The biological actions of LPA are mediated by at least six G protein-coupled receptors known as LPAR1-6. Given that LPAR1-3 were among the first LPARs identified, the majority of research efforts have focused on understanding their biology. This review provides an in-depth discussion of LPAR5, which has recently emerged as a key player in regulating normal intestinal homeostasis and modulating pathological conditions such as pain, itch, inflammatory diseases, and cancer. We also present a chronological overview of the efforts made to develop compounds that target LPAR5 for use as tool compounds to probe or validate LPAR5 biology and therapeutic agents for the treatment of inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Mélanie A Dacheux
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States of America
| | - Derek D Norman
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States of America
| | - Gábor J Tigyi
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States of America
| | - Sue Chin Lee
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States of America.
| |
Collapse
|
4
|
Mathew D, Torres RM. Lysophosphatidic Acid Is an Inflammatory Lipid Exploited by Cancers for Immune Evasion via Mechanisms Similar and Distinct From CTLA-4 and PD-1. Front Immunol 2021; 11:531910. [PMID: 33584637 PMCID: PMC7873449 DOI: 10.3389/fimmu.2020.531910] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Immunological tolerance has evolved to curtail immune responses against self-antigens and prevent autoimmunity. One mechanism that contributes to immunological tolerance is the expression of inhibitory receptors by lymphocytes that signal to dampen immune responses during the course of an infection and to prevent immune-mediated collateral damage to the host. The understanding that tumors exploit these physiological mechanisms to avoid elimination has led to remarkable, but limited, success in the treatment of cancer through the use of biologics that interfere with the ability of cancers to suppress immune function. This therapy, based on the understanding of how T lymphocytes are normally activated and suppressed, has led to the development of therapeutic blocking antibodies, referred to as immune checkpoint blockade, which either directly or indirectly promote the activation of CD8 T cells to eradicate cancer. Here, we highlight the distinct signaling mechanisms, timing and location of inhibition used by the CTLA-4 and PD-1 inhibitory receptors compared to a novel inhibitory signaling axis comprised of the bioactive lipid, lysophosphatidic acid (LPA), signaling via the LPA5 receptor expressed by CD8 T cells. Importantly, abundant evidence indicates that an LPA-LPA5 signaling axis is also exploited by diverse cancers to suppress T cell activation and function. Clearly, a thorough molecular and biochemical understanding of how diverse T cell inhibitory receptors signal to suppress T cell antigen receptor signaling and function will be important to inform the choice of which complimentary checkpoint blockade modalities might be used for a given cancer.
Collapse
Affiliation(s)
| | - Raul M. Torres
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
5
|
Dronkers E, Wauters MMM, Goumans MJ, Smits AM. Epicardial TGFβ and BMP Signaling in Cardiac Regeneration: What Lesson Can We Learn from the Developing Heart? Biomolecules 2020; 10:biom10030404. [PMID: 32150964 PMCID: PMC7175296 DOI: 10.3390/biom10030404] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/31/2022] Open
Abstract
The epicardium, the outer layer of the heart, has been of interest in cardiac research due to its vital role in the developing and diseased heart. During development, epicardial cells are active and supply cells and paracrine cues to the myocardium. In the injured adult heart, the epicardium is re-activated and recapitulates embryonic behavior that is essential for a proper repair response. Two indispensable processes for epicardial contribution to heart tissue formation are epithelial to mesenchymal transition (EMT), and tissue invasion. One of the key groups of cytokines regulating both EMT and invasion is the transforming growth factor β (TGFβ) family, including TGFβ and Bone Morphogenetic Protein (BMP). Abundant research has been performed to understand the role of TGFβ family signaling in the developing epicardium. However, less is known about signaling in the adult epicardium. This review provides an overview of the current knowledge on the role of TGFβ in epicardial behavior both in the development and in the repair of the heart. We aim to describe the presence of involved ligands and receptors to establish if and when signaling can occur. Finally, we discuss potential targets to improve the epicardial contribution to cardiac repair as a starting point for future investigation.
Collapse
|
6
|
Zhou Y, Little PJ, Ta HT, Xu S, Kamato D. Lysophosphatidic acid and its receptors: pharmacology and therapeutic potential in atherosclerosis and vascular disease. Pharmacol Ther 2019; 204:107404. [DOI: 10.1016/j.pharmthera.2019.107404] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 08/21/2019] [Indexed: 02/06/2023]
|
7
|
Main KA, Mikelis CM, Doçi CL. In Vitro Wound Healing Assays to Investigate Epidermal Migration. Methods Mol Biol 2019; 2109:147-154. [PMID: 31123996 DOI: 10.1007/7651_2019_235] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Re-epithelialization after cutaneous injury is a complex and multifaceted process that incorporates numerous cellular components interacting in a myriad of pathways. One of the most crucial aspects of this process is the initiation of keratinocyte migration to fill the wound bed. Re-epithelialization involves both the individual and collective movement of epidermal cells under the control of integrated signaling paradigms. It is therefore essential to develop a simple methodology to dissect the basic movement of epidermal cells in vitro. Scratch assays are relatively simple experiments in which a single layer of cells are plated onto a prepared dish with multiple furrows created in the cell bed. The resulting cellular migration to fill the wound bed can then be imaged and processed quantitatively to investigate migration rates and other factors of interest. Here, we provide important adaptations to the classic scratch assay to make it a robust, reproducible, and quantitative tool for the evaluation of epidermal cell migration.
Collapse
Affiliation(s)
- Kegan A Main
- Department of Biology, College of Arts and Sciences, Marian University Indianapolis, Indianapolis, IN, USA
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Colleen L Doçi
- Department of Biology, College of Arts and Sciences, Marian University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
8
|
Stuelten CH, Lee RM, Losert W, Parent CA. Lysophosphatidic acid regulates the motility of MCF10CA1a breast cancer cell sheets via two opposing signaling pathways. Cell Signal 2018; 45:1-11. [PMID: 29337044 PMCID: PMC5845779 DOI: 10.1016/j.cellsig.2018.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/22/2017] [Accepted: 01/07/2018] [Indexed: 01/31/2023]
Abstract
Aberrant cell migration leads to the dispersal of malignant cells. The ubiquitous lipid mediator lysophosphatidic acid (LPA) modulates cell migration and is implicated in tumor progression. Yet, the signaling cascades that regulate LPA's effect on cell motility remain unclear. Using time-lapse imaging and quantitative analyses, we studied the role of signaling cascades that act downstream of LPA on the motility of MCF10CA1a breast cancer cells. We found that LPA alters cell motility via two major signaling pathways. The Rho/ROCK signaling cascade is the predominant pathway that increases E-Cadherin containing cell-cell adhesions and cortical arrangement of actomyosin to promote slow, directional, spatially coherent and temporally consistent movement. In contrast, Gαi/o- and Gαq/11-dependent signaling cascades lessen directionality and support the independent movement of cells. The net effect of LPA on breast cancer cell migration therefore results from the integrated signaling activity of the Rho/ROCK and Gαi/o- and Gαq/11-dependent pathways, thus allowing for a dynamic migratory response to changes in the cellular or microenvironmental context.
Collapse
Affiliation(s)
- Christina H Stuelten
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States.
| | - Rachel M Lee
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States; Department of Physics, Physical Sciences Complex, University of Maryland, College Park, MD, United States
| | - Wolfgang Losert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States; Department of Physics, Physical Sciences Complex, University of Maryland, College Park, MD, United States
| | - Carole A Parent
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States; Department of Pharmacology, Michigan Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI..
| |
Collapse
|
9
|
Lysophosphatidic acid (LPA) as a pro-fibrotic and pro-oncogenic factor: a pivotal target to improve the radiotherapy therapeutic index. Oncotarget 2018; 8:43543-43554. [PMID: 28402936 PMCID: PMC5522168 DOI: 10.18632/oncotarget.16672] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/08/2017] [Indexed: 12/21/2022] Open
Abstract
Radiation-induced fibrosis is widely considered as a common but forsaken phenomenon that can lead to clinical sequela and possibly vital impairments. Lysophosphatidic acid is a bioactive lipid involved in fibrosis and probably in radiation-induced fibrosis as suggested in recent studies. Lysophosphatidic acid is also a well-described pro-oncogenic factor, involved in carcinogenesis processes (proliferation, survival, angiogenesis, invasion, migration). The present review highlights and summarizes the links between lysophosphatidic acid and radiation-induced fibrosis, lysophosphatidic acid and radioresistance, and proposes lysophosphatidic acid as a potential central actor of the radiotherapy therapeutic index. Besides, we hypothesize that following radiotherapy, the newly formed tumour micro-environment, with increased extracellular matrix and increased lysophosphatidic acid levels, is a favourable ground to metastasis development. Lysophosphatidic acid could therefore be an exciting therapeutic target, minimizing radio-toxicities and radio-resistance effects.
Collapse
|
10
|
Thorlakson HH, Engen SA, Schreurs O, Schenck K, Blix IJS. Lysophosphatidic acid induces expression of genes in human oral keratinocytes involved in wound healing. Arch Oral Biol 2017; 80:153-159. [DOI: 10.1016/j.archoralbio.2017.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 10/19/2022]
|
11
|
Doçi CL, Mikelis CM, Callejas-Valera JL, Hansen KK, Molinolo AA, Inoue A, Offermanns S, Gutkind JS. Epidermal loss of Gαq confers a migratory and differentiation defect in keratinocytes. PLoS One 2017; 12:e0173692. [PMID: 28301547 PMCID: PMC5354386 DOI: 10.1371/journal.pone.0173692] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 02/24/2017] [Indexed: 12/04/2022] Open
Abstract
G-protein coupled receptors (GPCRs), which activate heterotrimeric G proteins, are an essential class of transmembrane receptors that are responsible for a myriad of signaling events in normal and pathologic conditions. Two members of the G protein family, Gαq and Gα11, activate one of the main GPCR pathways and function as oncogenes by integrating mitogen-stimulated signaling cascades that are active under malignant conditions. Recently, it has been shown that targeted deletion of Gα11 and Gαq from endothelial cells impairs the Rho-mediated formation of focal adherens junctions, suggesting that Gα11/q signaling may also play a significant role in cytoskeletal-mediated cellular responses in epithelial cells. Indeed, combined deletion of Gα11 and Gαq confers a significant migratory defect in keratinocytes that delays cutaneous wound healing in an in vivo setting. This delay can be attributed to a defect during the reepithelialization phase due to significantly attenuated migratory capacity of Gαq-null keratinocytes under combined Gα11 deficiency. In fact, cells lacking Gα11/q demonstrate a severely reduced ability to respond to mitogenic and migratory signals in the microenvironment, leading to inappropriate and premature terminal differentiation. These results suggest that Gα11/q signaling pathways may be critical for integrating mitogenic signals and cytoskeletal function to achieve normal physiological responses. Emergence of a malignant phenotype may therefore arise from both under- and overexpression of Gα11/q signaling, implicating its upstream regulation as a potential therapeutic target in a host of pathologic conditions.
Collapse
Affiliation(s)
- Colleen L. Doçi
- College of Arts and Sciences, Marian University Indianapolis, Indianapolis, Indiana, United States of America
- * E-mail:
| | - Constantinos M. Mikelis
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, United States of America
| | - Juan Luis Callejas-Valera
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Karina K. Hansen
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alfredo A. Molinolo
- Department of Pathology, University of California San Diego, La Jolla, California, United States of America
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
- PRESTO, Japan Science and Technology Agency (JST), Kawaguchi, Saitama, Japan
| | - Stefan Offermanns
- Department of Pharmacology, Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - J. Silvio Gutkind
- Department of Pharmacology and Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
12
|
Thorlakson HH, Schreurs O, Schenck K, Blix IJS. Lysophosphatidic acid regulates adhesion molecules and enhances migration of human oral keratinocytes. Eur J Oral Sci 2016; 124:164-71. [PMID: 26913569 DOI: 10.1111/eos.12255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2016] [Indexed: 12/20/2022]
Abstract
Oral keratinocytes are connected via cell-to-cell adhesions to protect underlying tissues from physical and bacterial damage. Lysophosphatidic acids (LPAs) are a family of phospholipid mediators that have the ability to regulate gene expression, cytoskeletal rearrangement, and cytokine/chemokine secretion, which mediate proliferation, migration, and differentiation. Several forms of LPA are found in saliva and gingival crevicular fluid, but it is unknown how they affect human oral keratinocytes (HOK). The aim of the present study was therefore to examine how different LPA forms affect the expression of adhesion molecules and the migration and proliferation of HOK. Keratinocytes were isolated from gingival biopsies obtained from healthy donors and challenged with different forms of LPA. Quantitative real-time RT-PCR, immunocytochemistry, and flow cytometry were used to analyze the expression of adhesion molecules. Migration and proliferation assays were performed. Lysophosphatidic acids strongly promoted expression of E-cadherin and occludin mRNAs and translocation of E-cadherin protein from the cytoplasm to the membrane. Occludin and claudin-1 proteins were up-regulated by LPA. Migration of HOK in culture was increased, but proliferation was reduced, by the addition of LPA. This indicates that LPA can have a role in the regulation of the oral epithelial barrier by increasing the expression of adhesion molecules of HOK, by promotion of migration and by inhibition of proliferation.
Collapse
Affiliation(s)
- Hong H Thorlakson
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.,Department of Periodontology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Olav Schreurs
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Karl Schenck
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Inger J S Blix
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.,Department of Periodontology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
13
|
Nan L, Wei J, Jacko AM, Culley MK, Zhao J, Natarajan V, Ma H, Zhao Y. Cross-talk between lysophosphatidic acid receptor 1 and tropomyosin receptor kinase A promotes lung epithelial cell migration. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:229-35. [PMID: 26597701 DOI: 10.1016/j.bbamcr.2015.11.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/28/2015] [Accepted: 11/16/2015] [Indexed: 02/02/2023]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lysophospholipid, which plays a crucial role in the regulation of cell proliferation, migration, and differentiation. LPA exerts its biological effects mainly through binding to cell-surface LPA receptors (LPA1-6), which belong to the G protein-coupled receptor (GPCR) family. Recent studies suggest that cross-talk between receptor tyrosine kinases (RTKs) and GPCRs modulates GPCRs-mediated signaling. Tropomyosin receptor kinase A (TrkA) is a RTK, which mediates nerve growth factor (NGF)-induced biological functions including cell migration in neuronal and non-neuronal cells. Here, we show LPA1 transactivation of TrkA in murine lung epithelial cells (MLE12). LPA induced tyrosine phosphorylation of TrkA in both time- and dose-dependent manners. Down-regulation of LPA1 by siRNA transfection attenuated LPA-induced phosphorylation of TrkA, suggesting a cross-talk between LPA1 and TrkA. To investigate the molecular regulation of the cross-talk, we focused on the interaction between LPA1 and TrkA. We found that LPA induced interaction between LPA1 and TrkA. The LPA1/TrkA complex was localized on the plasma membrane and in the cytoplasm. The C-terminus of LPA1 was identified as the binding site for TrkA. Inhibition of TrkA attenuated LPA-induced phosphorylation of TrkA and LPA1 internalization, as well as lung epithelial cell migration. These studies provide a molecular mechanism for the transactivation of TrkA by LPA, and suggest that the cross-talk between LPA1 and TrkA regulates LPA-induced receptor internalization and lung epithelial cell migration.
Collapse
Affiliation(s)
- Ling Nan
- Department of Anesthesia, First Hospital of Jilin University, Changchun, China; Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jianxin Wei
- Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Anastasia M Jacko
- Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Miranda K Culley
- Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jing Zhao
- Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, United States
| | - Haichun Ma
- Department of Anesthesia, First Hospital of Jilin University, Changchun, China
| | - Yutong Zhao
- Department of Medicine, Acute Lung Injury Center of Excellence, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
14
|
Ohashi T, Yamamoto T. Antifibrotic effect of lysophosphatidic acid receptors LPA1and LPA3antagonist on experimental murine scleroderma induced by bleomycin. Exp Dermatol 2015; 24:698-702. [DOI: 10.1111/exd.12752] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Takenobu Ohashi
- Department of Dermatology; Fukushima Medical University; Fukushima Japan
| | - Toshiyuki Yamamoto
- Department of Dermatology; Fukushima Medical University; Fukushima Japan
| |
Collapse
|
15
|
KANG SANGJIN, HAN JUHEE, SONG SEUNGYONG, KIM WONSERK, SHIN SOYOUNG, KIM JIHYE, AHN HYOSUN, JEONG JINHYUN, HWANG SUNGJOO, SUNG JONGHYUK. Lysophosphatidic acid increases the proliferation and migration of adipose-derived stem cells via the generation of reactive oxygen species. Mol Med Rep 2015; 12:5203-10. [DOI: 10.3892/mmr.2015.4023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 06/15/2015] [Indexed: 11/06/2022] Open
|
16
|
Lysophosphatidic acid promotes cell migration through STIM1- and Orai1-mediated Ca2+(i) mobilization and NFAT2 activation. J Invest Dermatol 2012; 133:793-802. [PMID: 23096711 PMCID: PMC3572452 DOI: 10.1038/jid.2012.370] [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] [Indexed: 01/03/2023]
Abstract
Lysophosphatidic acid (LPA) enhances cell migration and promotes wound healing in vivo, but the intracellular signaling pathways regulating these processes remain incompletely understood. Here we investigated the involvement of agonist-induced Ca2+ entry and STIM1 and Orai1 proteins in regulating nuclear factor of activated T cell (NFAT) signaling and LPA-induced keratinocyte cell motility. As monitored by Fluo-4 imaging, stimulation with 10 μℳ LPA in 60 μℳ Ca2+o evoked Ca2+i transients owing to store release, whereas addition of LPA in physiological 1.2 mℳ Ca2+o triggered store release coupled to extracellular Ca2+ entry. Store-operated Ca2+ entry (SOCE) was blocked by the SOCE inhibitor diethylstilbestrol (DES), STIM1 silencing using RNA interference (RNAi), and expression of dominant/negative Orai1R91W. LPA induced significant NFAT activation as monitored by nuclear translocation of green fluorescent protein-tagged NFAT2 and a luciferase reporter assay, which was impaired by DES, expression of Orai1R91W, and inhibition of calcineurin using cyclosporin A (CsA). By using chemotactic migration assays, LPA-induced cell motility was significantly impaired by STIM1, CsA, and NFAT2 knockdown using RNAi. These data indicate that in conditions relevant to epidermal wound healing, LPA induces SOCE and NFAT activation through Orai1 channels and promotes cell migration through a calcineurin/NFAT2-dependent pathway.
Collapse
|
17
|
Gene expression of the lysophosphatidic acid receptor 1 is a target of transforming growth factor beta. Oncogene 2012; 32:3198-206. [PMID: 22824789 PMCID: PMC3480976 DOI: 10.1038/onc.2012.325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The lysophosphatidic acid (LPA) receptor LPA1/Edg2 is the first identified LPA receptor. Although its wide tissue distribution and biological functions have been well studied, little is known about how LPA1 is transcriptionally regulated. In the current study, we showed that LPA1 is a physiological target of transforming growth factor beta (TGFβ)-mediated repression. In both normal and neoplastic cells, TGFβ inhibits LPA1 promoter activity, LPA1 mRNA expression, and LPA1-dependent chemotaxis and tumor cell invasion. Knockdown of the TGFβ intracellular effector Smad3 or Smad4 with lentivirally transduced shRNA relieved these inhibitory effects of TGFβ. Interestingly, the LPA1 promoter contains two potential TGFβ inhibitory elements (TIEs), each consisting of a Smad binding site and an adjacent E2F4/5 element, structurally similar to the TIE found on the promoter of the well-defined TGFβ target gene c-myc. Deletion and point mutation analyses indicate that the distal TIE located at 401 bp from the transcription initiation site, is required for TGFβ repression of the LPA1 promoter. A DNA pull-down assay showed that the -401 TIE was capable of binding Samd3 and E2F4 in TGFβ-treated cells. TGFβ-induced binding of the Smad complex to the native -401 TIE sequence of the LPA1 gene promoter was further verified by chromatin immunoprecipitation assays. We therefore identified a novel role of TGFβ in the control of LPA1 expression and LPA1-coupled biological functions, adding LPA1 to the list of TGFβ-repressed target genes.
Collapse
|
18
|
Jeon ES, Kim JH, Ryu H, Kim EK. Lysophosphatidic acid activates TGFBIp expression in human corneal fibroblasts through a TGF-β1-dependent pathway. Cell Signal 2012; 24:1241-50. [DOI: 10.1016/j.cellsig.2012.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 02/14/2012] [Accepted: 02/14/2012] [Indexed: 11/24/2022]
|
19
|
Somatostatin inhibits cell migration and reduces cell counts of human keratinocytes and delays epidermal wound healing in an ex vivo wound model. PLoS One 2011; 6:e19740. [PMID: 21589940 PMCID: PMC3092774 DOI: 10.1371/journal.pone.0019740] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 04/14/2011] [Indexed: 12/26/2022] Open
Abstract
The peptide hormone somatostatin (SST) and its five G protein-coupled receptors
(SSTR1-5) were described to be present in the skin, but their cutaneous
function(s) and skin-specific signalling mechanisms are widely unknown. By using
receptor specific agonists we show here that the SSTRs expressed in
keratinocytes are functionally coupled to the inhibition of adenylate cyclase.
In addition, treatment with SSTR4 and SSTR5/1 specific agonists significantly
influences the MAP kinase signalling pathway. As epidermal hormone receptors in
general are known to regulate re-epithelialization following skin injury, we
investigated the effect of SST on cell counts and migration of human
keratinocytes. Our results demonstrate a significant inhibition of cell
migration and reduction of cell counts by SST. We do not observe an effect on
apoptosis and necrosis. Analysis of signalling pathways showed that somatostatin
inhibits cell migration independent of its effect on cAMP. Migrating
keratinocytes treated with SST show altered cytoskeleton dynamics with delayed
lamellipodia formation. Furthermore, the activity of the small GTPase Rac1 is
diminished, providing evidence for the control of the actin cytoskeleton by
somatostatin receptors in keratinocytes. While activation of all receptors leads
to redundant effects on cell migration, only treatment with a SSTR5/1 specific
agonist resulted in decreased cell counts. In accordance with reduced cell
counts and impaired migration we observe delayed re-epithelialization in an
ex vivo wound healing model. Consequently, our experiments
suggest SST as a negative regulator of epidermal wound healing.
Collapse
|
20
|
Yin Z, Carbone LD, Gotoh M, Postlethwaite A, Bolen AL, Tigyi GJ, Murakami-Murofushi K, Watsky MA. Lysophosphatidic acid-activated Cl- current activity in human systemic sclerosis skin fibroblasts. Rheumatology (Oxford) 2010; 49:2290-7. [PMID: 20823096 DOI: 10.1093/rheumatology/keq260] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVES SSc (scleroderma) is an often fatal disease characterized by widespread tissue fibrosis. Fibroblasts play a key role in SSc-associated fibrosis. This study was designed to determine: (i) whether fibroblasts isolated from skin of patients with SSc have increased lysophosphatidic acid-activated Cl- current (IClLPA) activity vs healthy controls; (ii) whether myofibroblast differentiation is involved in SSc skin fibrosis; and (iii) whether SSc fibroblasts have different proliferation rates vs controls. METHODS Skin biopsies were taken from involved and uninvolved skin of SSc patients and controls. Whole-cell perforated patch-clamping was used to measure IClLPA activity in fibroblasts isolated and cultured from these biopsies. Western blotting was used to measure α-smooth muscle actin (α-SMA). Proliferation was measured using a colorimetric assay. RESULTS Fibroblasts cultured from SSc skin show significantly increased IClLPA activity following LPA exposure compared with control skin fibroblasts. α-SMA protein was significantly increased in cultured SSc skin fibroblasts vs controls. No significant differences in proliferation rates were found. CONCLUSIONS Elevated IClLPA activity is a hallmark of SSc skin fibroblasts. Blocking IClLPA activation may be a new therapeutic approach for treating SSc-associated fibrosis.
Collapse
Affiliation(s)
- Zhaohong Yin
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Ave., Memphis, TN 38163, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Lysophosphatidic Acid Upregulates Laminin-332 Expression during A431 Cell Colony Dispersal. JOURNAL OF ONCOLOGY 2010; 2010. [PMID: 20862207 PMCID: PMC2938436 DOI: 10.1155/2010/107075] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 06/11/2010] [Accepted: 07/02/2010] [Indexed: 01/01/2023]
Abstract
Lysophosphatidic acid (LPA) is a bioactive phospholipid that affects various biological functions, such as cell proliferation, migration, survival, wound healing, and tumor invasion through LPA receptors. Previously, we reported that LPA induces A431 colony dispersal, accompanied by disruption of cell-cell contacts and cell migration. However, it remains unclear how LPA affects cell migration and gene expression during A431 colony dispersal. In this paper, we performed cDNA microarray analysis to investigate this question by comparing gene expression between untreated and LPA-treated A431 cells. Interestingly, these results revealed that LPA treatment upregulates several TGF-β1 target genes, including laminin-332 (Ln-332) components (α3, β3, and γ2 chains). Western blot analysis also showed that LPA increased phosphorylation of Smad2, an event that is carried out by TGF-β1 interactions. Among the genes upregulated, we further addressed the role of Ln-332. Real-time PCR analysis confirmed the transcriptional upregulation of all α3, β3, and γ2 chains of Ln-332 by LPA, corresponding to the protein level increases revealed by western blot. Further, the addition of anti-Ln-332 antibody prevented LPA-treated A431 colonies from dispersing. Taken together, our results suggest that LPA-induced Ln-332 plays a significant role in migration of individual cells from A431 colonies.
Collapse
|
22
|
Watsky MA, Weber KT, Sun Y, Postlethwaite A. New insights into the mechanism of fibroblast to myofibroblast transformation and associated pathologies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 282:165-92. [PMID: 20630469 DOI: 10.1016/s1937-6448(10)82004-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Myofibroblasts are a differentiated cell type essential for wound healing, participating in tissue remodeling following insult. Myofibroblasts are typically activated fibroblasts, although they can also be derived from other cell types, including epithelial cells, endothelial cells, and mononuclear cells. In most organ systems, cell signals initiated following tissue-specific insult or during the metastatic process lead to differentiation of fibroblasts or other precursor cells to the myofibroblast phenotype. In addition to their beneficial and necessary role in wound healing, myofibroblasts also contribute to a number of pathologies, primarily fibrotic processes and tumor invasiveness. This review explores both traditional and nontraditional concepts of myofibroblast differentiation in the cornea, skin, heart, and other tissues, as well as some of the pathologies associated with myofibroblast activities.
Collapse
Affiliation(s)
- Mitchell A Watsky
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | | | | |
Collapse
|
23
|
Influences of opioids and nanoparticles on in vitro wound healing models. Eur J Pharm Biopharm 2009; 73:34-42. [DOI: 10.1016/j.ejpb.2009.03.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 03/18/2009] [Accepted: 03/25/2009] [Indexed: 11/18/2022]
|
24
|
Lichte K, Rossi R, Danneberg K, Braak MT, Kürschner U, Jakobs KH, Kleuser B, Heringdorf DMZ. Lysophospholipid Receptor-Mediated Calcium Signaling in Human Keratinocytes. J Invest Dermatol 2008; 128:1487-98. [DOI: 10.1038/sj.jid.5701207] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
25
|
Abstract
The mechanisms of agonist-induced calcium entry (ACE) following depletion of intracellular calcium stores have not been fully established. We report here that calcium-independent phospholipase A (iPLA2) is required for robust Ca2+ entry in HaCaT keratinocytes following ATP or UTP stimulation. Lysophosphatidic acid (LPA), an unrelated agonist, evoked Ca2+ release without inducing robust Ca2+ entry. Both LPA and UTP induced the redistribution of STIM1 into puncta which localized to regions near or at the plasma membrane, as well as within the cytoplasm. Plasma membrane-associated STIM1 remained high for up to 10 min after UTP stimulation, whereas it had returned almost to baseline by that time point in LPA-stimulated cells. This correlated with faster reloading of the endoplasmic reticulum Ca2+ stores in LPA treated cells. Thus by differentially regulating store-refilling after agonist-mediated depletion, LPA and UTP may exert distinct effects on the duration of STIM1 localization at the plasma membrane, and thus, on the magnitude and duration of ACE.
Collapse
Affiliation(s)
- Kehinde Ross
- Dermatological Sciences, Institute of Cellular Medicine, Medical School, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | | | | |
Collapse
|
26
|
van Meeteren LA, Moolenaar WH. Regulation and biological activities of the autotaxin-LPA axis. Prog Lipid Res 2007; 46:145-60. [PMID: 17459484 DOI: 10.1016/j.plipres.2007.02.001] [Citation(s) in RCA: 292] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2007] [Revised: 02/20/2007] [Accepted: 02/23/2007] [Indexed: 12/22/2022]
Abstract
Autotaxin (ATX), or nucleotide pyrophosphatase/phosphodiesterase 2 (NPP2), is an exo-enzyme originally identified as a tumor cell autocrine motility factor. ATX is unique among the NPPs in that it primarily functions as a lysophospholipase D, converting lysophosphatidylcholine into the lipid mediator lysophosphatidic acid (LPA). LPA acts on specific G protein-coupled receptors to elicit a wide range of cellular responses, ranging from cell proliferation and migration to neurite remodeling and cytokine production. While LPA signaling has been studied extensively over the last decade, we are only now beginning to explore the properties and biological importance of ATX as the major LPA-producing phospholipase. In this review, we highlight recent advances in our understanding of the ATX-LPA axis, giving first an update on LPA action and then focusing on ATX, in particular its regulation, its link to cancer and its vital role in vascular development.
Collapse
Affiliation(s)
- Laurens A van Meeteren
- Division of Cellular Biochemistry, Centre for Biomedical Genetics, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, The Netherlands
| | | |
Collapse
|
27
|
Wu WS, Tsai RK, Chang CH, Wang S, Wu JR, Chang YX. Reactive Oxygen Species Mediated Sustained Activation of Protein Kinase C α and Extracellular Signal-Regulated Kinase for Migration of Human Hepatoma Cell Hepg2. Mol Cancer Res 2006; 4:747-58. [PMID: 17050668 DOI: 10.1158/1541-7786.mcr-06-0096] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA) can trigger growth inhibition, epithelial-mesenchymal transition (EMT)-like cell scattering, and migration of hepatoma cells HepG2 in a protein kinase C-alpha (PKC-alpha)-dependent manner. Saikosaponin a, an ingredient of antitumorigenic Chinese herb Sho-Saiko-to, inhibited cell growth but did not induce EMT-like cell scattering and cell migration of HepG2. Saikosaponin a and TPA induced transient (for 30 minutes) and sustained (until 6 hours) phosphorylation of extracellular signal-regulated kinase (ERK), respectively. Generation of the reactive oxygen species (ROS) was induced by TPA, but not saikosaponin a, for 3 hours. As expected, scavengers of ROS, such as superoxide dismutase, catalase, and mannitol, and the thiol-containing antioxidant N-acetylcystein dramatically suppressed the TPA-triggered cell migration but not growth inhibition of HepG2. The generation of ROS induced by TPA was PKC, but not ERK, dependent. On the other hand, scavengers of ROS and N-acetylcystein also prevented PKC activation and ERK phosphorylation induced by TPA. On the transcriptional level, TPA can induce gene expression of integrins alpha5, alpha6, and beta1 and reduce gene expression of E-cahedrin in a PKC- and ROS-dependent manner. In conclusion, ROS play a central role in mediating TPA-triggered sustained PKC and ERK signaling for regulation of gene expression of integrins and E-cahedrin that are responsible for EMT and migration of HepG2.
Collapse
Affiliation(s)
- Wen-Sheng Wu
- Department of Medical Technology, Tzu Chi University, No. 701, Chung Yang Road, Section 3, Hualien 970, Taiwan.
| | | | | | | | | | | |
Collapse
|
28
|
Borzini P, Mazzucco L. Tissue regeneration and in loco administration of platelet derivatives: clinical outcome, heterogeneous products, and heterogeneity of the effector mechanisms. Transfusion 2005; 45:1759-67. [PMID: 16271101 DOI: 10.1111/j.1537-2995.2005.00600.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND In loco administration of platelet (PLT) derivatives is a relatively new auxiliary treatment for tissue regeneration to be hastened. Enthusiastic reports are faced by more critical ones. The more obvious rationale for the in vivo administration of PLT derivatives resides in their growth factor content. STUDY DESIGN AND METHODS The relevant literature was systematically reviewed. Close scrutiny of the technical details was carried out to find out the procedural differences accounting for conflicting results. RESULTS An impressively vast heterogeneity of conduct was found in both in vitro and in vivo studies. Major outcome-affecting variables were recognized such as those associated with PLT preparation; growth factor measurement; proliferation test; dose, timing, and administration of the PLT derivatives; study design; and primary endpoints. CONCLUSIONS So many variables were found making standardization or confrontation of the in vitro and the in vivo studies barely conceivable or manageable. The mechanisms of action are very complex. The attribution of tissue regeneration capacity of PLT derivatives solely to the PLT-derived growth factors is simplistic. The results obtained through in vitro experiments are indicative for general mechanisms. Their simplistic hold to the complex in vivo environment may be misleading.
Collapse
Affiliation(s)
- Piero Borzini
- Department of Hematology and Transfusion Medicine, Transfusion Medicine Service, SS Antonio and Biagio Hospital, Alessandria, Italy.
| | | |
Collapse
|
29
|
Mazereeuw-Hautier J, Gres S, Fanguin M, Cariven C, Fauvel J, Perret B, Chap H, Salles JP, Saulnier-Blache JS. Production of lysophosphatidic acid in blister fluid: involvement of a lysophospholipase D activity. J Invest Dermatol 2005; 125:421-7. [PMID: 16117781 PMCID: PMC1885457 DOI: 10.1111/j.0022-202x.2005.23855.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Lysophosphatidic acid (LPA) is present in abundance in serum resulting from platelet activation and is also found in other biological fluids. LPA controls numerous cellular responses and plays a role in specific functions such as wound healing, especially in the skin. Nevertheless, its presence in the skin has never been investigated. Since re-epithelialization occurs after blister rupture, we tested the presence of endogenous LPA in blister fluid and investigated a possible mechanism for its biosynthesis and biological functions. Using a radioenzymatic assay, LPA was detected in 33 blister fluids originating from 24 bullous dermatoses, and at higher concentrations than in plasma. In parallel, blister fluids contained a lysophospholipase D (LPLD) activity but no detectable phospholipase A2 activity. The expressions of the LPLD autotaxin (ATX) and of LPA1-receptor (LPA1-R) were greatly increased in blister skin when compared with normal skin. Finally, LPA was found to have a positive effect on the migration of cultured keratinocytes. These results show that LPA is present in blister fluid synthesized by the LPLD ATX. Due to its ability to enhance keratinocyte migration, LPA in blister fluid could, via the LPA1-R, play an important role in re-epithelialization occurring after blister rupture.
Collapse
|