1
|
Sporkova A, Ghosh S, Al-Hasani J, Hecker M. Lin11-Isl1-Mec3 Domain Proteins as Mechanotransducers in Endothelial and Vascular Smooth Muscle Cells. Front Physiol 2021; 12:769321. [PMID: 34867475 PMCID: PMC8640458 DOI: 10.3389/fphys.2021.769321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/26/2021] [Indexed: 11/13/2022] Open
Abstract
Arterial hypertension is the leading risk factor for cardiovascular morbidity and mortality worldwide. However, little is known about the cellular mechanisms underlying it. In small arteries and arterioles, a chronic increase in blood pressure raises wall tension and hence stretches, namely, the medial vascular smooth muscle cells (VSMC) but also endothelial cell (EC) to cell contacts. Initially compensated by an increase in vascular tone, the continuous biomechanical strain causes a prominent change in gene expression in both cell types, frequently driving an arterial inward remodeling process that ultimately results in a reduction in lumen diameter, stiffening of the vessel wall, and fixation of blood pressure, namely, diastolic blood pressure, at the elevated level. Sensing and propagation of this supraphysiological stretch into the nucleus of VSMC and EC therefore seems to be a crucial step in the initiation and advancement of hypertension-induced arterial remodeling. Focal adhesions (FA) represent an important interface between the extracellular matrix and Lin11-Isl1-Mec3 (LIM) domain-containing proteins, which can translocate from the FA into the nucleus where they affect gene expression. The varying biomechanical cues to which vascular cells are exposed can thus be rapidly and specifically propagated to the nucleus. Zyxin was the first protein described with such mechanotransducing properties. It comprises 3 C-terminal LIM domains, a leucine-rich nuclear export signal, and N-terminal features that support its association with the actin cytoskeleton. In the cytoplasm, zyxin promotes actin assembly and organization as well as cell motility. In EC, zyxin acts as a transcription factor, whereas in VSMC, it has a less direct effect on mechanosensitive gene expression. In terms of homology and structural features, lipoma preferred partner is the nearest relative of zyxin among the LIM domain proteins. It is almost exclusively expressed by smooth muscle cells in the adult, resides like zyxin at FA but seems to affect mechanosensitive gene expression indirectly, possibly via altering cortical actin dynamics. Here, we highlight what is currently known about the role of these LIM domain proteins in mechanosensing and transduction in vascular cells.
Collapse
Affiliation(s)
- Alexandra Sporkova
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Subhajit Ghosh
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany
| | - Jaafar Al-Hasani
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site, Heidelberg/Mannheim, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research) Partner Site, Heidelberg/Mannheim, Germany
| |
Collapse
|
2
|
Ramelli SC, Comer BS, McLendon JM, Sandy LL, Ferretti AP, Barrington R, Sparks J, Matar M, Fewell J, Gerthoffer WT. Nanoparticle Delivery of Anti-inflammatory LNA Oligonucleotides Prevents Airway Inflammation in a HDM Model of Asthma. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:1000-1014. [PMID: 32044723 PMCID: PMC7013130 DOI: 10.1016/j.omtn.2019.12.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/16/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023]
Abstract
To address the problem of poor asthma control due to drug resistance, an antisense oligonucleotide complementary to mmu-miR-145a-5p (antimiR-145) was tested in a house dust mite mouse model of mild/moderate asthma. miR-145 was targeted to reduce inflammation, regulate epithelial-mesenchymal transitions, and promote differentiation of structural cells. In addition, several chemical variations of a nontargeting oligonucleotide were tested to define sequence-dependent effects of the miRNA antagonist. After intravenous administration, oligonucleotides complexed with a pegylated cationic lipid nanoparticle distributed to most cells in the lung parenchyma but were not present in smooth muscle or the mucosal epithelium of the upper airways. Treatment with antimiR-145 and a nontargeting oligonucleotide both reduced eosinophilia, reduced obstructive airway remodeling, reduced mucosal metaplasia, and reduced CD68 immunoreactivity. Poly(A) RNA-seq verified that antimiR-145 increased levels of many miR-145 target transcripts. Genes upregulated in human asthma and the mouse model of asthma were downregulated by oligonucleotide treatments. However, both oligonucleotides significantly upregulated many genes of interferon signaling pathways. These results establish effective lung delivery and efficacy of locked nucleic acid/DNA oligonucleotides administered intravenously, and suggest that some of the beneficial effects of oligonucleotide therapy of lung inflammation may be due to normalization of interferon response pathways.
Collapse
Affiliation(s)
- Sabrina C Ramelli
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Brian S Comer
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Jared M McLendon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Lydia L Sandy
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Andrew P Ferretti
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, USA
| | - Robert Barrington
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, USA
| | - Jeff Sparks
- Celsion Corporation, 601 Genome Way, Huntsville, AL, USA
| | - Majed Matar
- Celsion Corporation, 601 Genome Way, Huntsville, AL, USA
| | - Jason Fewell
- Celsion Corporation, 601 Genome Way, Huntsville, AL, USA
| | - William T Gerthoffer
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA; Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, USA.
| |
Collapse
|
3
|
Nelms BD, Waldron L, Barrera LA, Weflen AW, Goettel JA, Guo G, Montgomery RK, Neutra MR, Breault DT, Snapper SB, Orkin SH, Bulyk ML, Huttenhower C, Lencer WI. CellMapper: rapid and accurate inference of gene expression in difficult-to-isolate cell types. Genome Biol 2016; 17:201. [PMID: 27687735 PMCID: PMC5043525 DOI: 10.1186/s13059-016-1062-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/13/2016] [Indexed: 02/25/2023] Open
Abstract
We present a sensitive approach to predict genes expressed selectively in specific cell types, by searching publicly available expression data for genes with a similar expression profile to known cell-specific markers. Our method, CellMapper, strongly outperforms previous computational algorithms to predict cell type-specific expression, especially for rare and difficult-to-isolate cell types. Furthermore, CellMapper makes accurate predictions for human brain cell types that have never been isolated, and can be rapidly applied to diverse cell types from many tissues. We demonstrate a clinically relevant application to prioritize candidate genes in disease susceptibility loci identified by GWAS.
Collapse
Affiliation(s)
- Bradlee D Nelms
- Division of Gastroenterology, Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA. .,Graduate Program in Biophysics, Harvard University, Cambridge, MA, 02138, USA.
| | - Levi Waldron
- City University of New York School of Public Health, New York, NY, 10027, USA
| | - Luis A Barrera
- Graduate Program in Biophysics, Harvard University, Cambridge, MA, 02138, USA.,Division of Genetics, Department of Medicine and Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew W Weflen
- Division of Gastroenterology, Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jeremy A Goettel
- Division of Gastroenterology, Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Guoji Guo
- Center of Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Zhejiang, 310058, People's Republic of China
| | - Robert K Montgomery
- Division of Gastroenterology, Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Marian R Neutra
- Division of Gastroenterology, Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA, 02115, USA
| | - David T Breault
- Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA, 02115, USA.,Division of Endocrinology, Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Scott B Snapper
- Division of Gastroenterology, Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA, 02115, USA.,Department of Gastroenterology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Stuart H Orkin
- Division of Hematology/Oncology and Harvard Stem Cell Institute, Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Martha L Bulyk
- Division of Genetics, Department of Medicine and Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Wayne I Lencer
- Division of Gastroenterology, Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA. .,Graduate Program in Biophysics, Harvard University, Cambridge, MA, 02138, USA. .,Harvard Digestive Diseases Center, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
4
|
Perisic L, Aldi S, Sun Y, Folkersen L, Razuvaev A, Roy J, Lengquist M, Åkesson S, Wheelock CE, Maegdefessel L, Gabrielsen A, Odeberg J, Hansson GK, Paulsson-Berne G, Hedin U. Gene expression signatures, pathways and networks in carotid atherosclerosis. J Intern Med 2016; 279:293-308. [PMID: 26620734 DOI: 10.1111/joim.12448] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Embolism from unstable atheromas in the carotid bifurcation is a major cause of stroke. Here, we analysed gene expression in endarterectomies from patients with symptomatic (S) and asymptomatic (AS) carotid stenosis to identify pathways linked to plaque instability. METHODS Microarrays were prepared from plaques (n = 127) and peripheral blood samples (n = 96) of S and AS patients. Gene set enrichment, pathway mapping and network analyses of differentially expressed genes were performed. RESULTS These studies revealed upregulation of haemoglobin metabolism (P = 2.20E-05) and bone resorption (P = 9.63E-04) in S patients. Analysis of subgroups of patients indicated enrichment of calcification and osteoblast differentiation in S patients on statins, as well as inflammation and apoptosis in plaques removed >1 month compared to <2 weeks after symptom. By prediction profiling, a panel of 30 genes, mostly transcription factors, discriminated between plaques from S versus AS patients with 78% accuracy. By meta-analysis, common gene networks associated with atherosclerosis mapped to hypoxia, chemokines, calcification, actin cytoskeleton and extracellular matrix. A set of dysregulated genes (LMOD1, SYNPO2, PLIN2 and PPBP) previously not described in atherosclerosis were identified from microarrays and validated by quantitative PCR and immunohistochemistry. CONCLUSIONS Our findings confirmed a central role for inflammation and proteases in plaque instability, and highlighted haemoglobin metabolism and bone resorption as important pathways. Subgroup analysis suggested prolonged inflammation following the symptoms of plaque instability and calcification as a possible stabilizing mechanism by statins. In addition, transcriptional regulation may play an important role in the determination of plaque phenotype. The results from this study will serve as a basis for further exploration of molecular signatures in carotid atherosclerosis.
Collapse
Affiliation(s)
- L Perisic
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - S Aldi
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Y Sun
- Translational Science Center, Personalized Healthcare and Biomarkers, R&D, Astra Zeneca, Stockholm, Sweden
| | - L Folkersen
- Department of Molecular Genetics, Novo Nordisk, Copenhagen, Denmark.,Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - A Razuvaev
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - J Roy
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - M Lengquist
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - S Åkesson
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - C E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - L Maegdefessel
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - A Gabrielsen
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - J Odeberg
- Department of Medicine, Karolinska Institute, Stockholm, Sweden.,Science for Life Laboratory, Department of Proteomics, School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | - G K Hansson
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - U Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
5
|
Murray IR, West CC, Hardy WR, James AW, Park TS, Nguyen A, Tawonsawatruk T, Lazzari L, Soo C, Péault B. Natural history of mesenchymal stem cells, from vessel walls to culture vessels. Cell Mol Life Sci 2014; 71:1353-74. [PMID: 24158496 PMCID: PMC11113613 DOI: 10.1007/s00018-013-1462-6] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 08/17/2013] [Accepted: 08/23/2013] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) can regenerate tissues by direct differentiation or indirectly by stimulating angiogenesis, limiting inflammation, and recruiting tissue-specific progenitor cells. MSCs emerge and multiply in long-term cultures of total cells from the bone marrow or multiple other organs. Such a derivation in vitro is simple and convenient, hence popular, but has long precluded understanding of the native identity, tissue distribution, frequency, and natural role of MSCs, which have been defined and validated exclusively in terms of surface marker expression and developmental potential in culture into bone, cartilage, and fat. Such simple, widely accepted criteria uniformly typify MSCs, even though some differences in potential exist, depending on tissue sources. Combined immunohistochemistry, flow cytometry, and cell culture have allowed tracking the artifactual cultured mesenchymal stem/stromal cells back to perivascular anatomical regions. Presently, both pericytes enveloping microvessels and adventitial cells surrounding larger arteries and veins have been described as possible MSC forerunners. While such a vascular association would explain why MSCs have been isolated from virtually all tissues tested, the origin of the MSCs grown from umbilical cord blood remains unknown. In fact, most aspects of the biology of perivascular MSCs are still obscure, from the emergence of these cells in the embryo to the molecular control of their activity in adult tissues. Such dark areas have not compromised intents to use these cells in clinical settings though, in which purified perivascular cells already exhibit decisive advantages over conventional MSCs, including purity, thorough characterization and, principally, total independence from in vitro culture. A growing body of experimental data is currently paving the way to the medical usage of autologous sorted perivascular cells for indications in which MSCs have been previously contemplated or actually used, such as bone regeneration and cardiovascular tissue repair.
Collapse
Affiliation(s)
- Iain R. Murray
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Christopher C. West
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Winters R. Hardy
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
- Indiana Center for Vascular Biology and Medicine, Indianapolis, USA
| | - Aaron W. James
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Tea Soon Park
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore, USA
| | - Alan Nguyen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Tulyapruek Tawonsawatruk
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Lorenza Lazzari
- Cell Factory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Departments of Surgery and Orthopedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Bruno Péault
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
- BHF Center for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Orthopedic Hospital Research Center and Broad Stem Cell Center, David Geffen School of Medicine, University of California, Los Angeles, USA
| |
Collapse
|
6
|
Cell Adhesion and Transcriptional Activity - Defining the Role of the Novel Protooncogene LPP. Transl Oncol 2011; 2:107-16. [PMID: 19701494 DOI: 10.1593/tlo.09112] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 02/20/2009] [Accepted: 02/25/2009] [Indexed: 12/13/2022] Open
Abstract
Integrating signals from the extracellular matrix through the cell surface into the nucleus is an essential feature of metazoan life. To date, many signal transducers known as shuttle proteins have been identified to act as both a cytoskeletal and a signaling protein. Among them, the most prominent representatives are zyxin and lipoma preferred (translocation) partner (LPP). These proteins belong to the LIM domain protein family and are associated with cell migration, proliferation, and transcription. LPP was first identified in benign human lipomas and was subsequently found to be overexpressed in human malignancies such as lung carcinoma, soft tissue sarcoma, and leukemia. This review portrays LPP in the context of human neoplasia based on a study of the literature to define its important role as a novel protooncogene in carcinogenesis.
Collapse
|
7
|
Linnemann A, van der Ven PFM, Vakeel P, Albinus B, Simonis D, Bendas G, Schenk JA, Micheel B, Kley RA, Fürst DO. The sarcomeric Z-disc component myopodin is a multiadapter protein that interacts with filamin and alpha-actinin. Eur J Cell Biol 2010; 89:681-92. [PMID: 20554076 DOI: 10.1016/j.ejcb.2010.04.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 04/15/2010] [Accepted: 04/16/2010] [Indexed: 11/28/2022] Open
Abstract
Here we introduce myopodin as a novel filamin C binding partner. Corroborative yeast two-hybrid and biochemical analyses indicate that the central part of myopodin that shows high homology to the closely related protein synaptopodin and that is common to all its currently known or predicted variants interacts with filamin C immunoglobulin-like domains 20-21. A detailed characterization of the previously described interaction between myopodin and alpha-actinin demonstrates for the first time that myopodin contains three independent alpha-actinin-binding sites. Newly developed myopodin-specific antibodies reveal expression at the earliest stages of in vitro differentiation of human skeletal muscle cells preceding the expression of sarcomeric alpha-actinin. Myopodin colocalizes with filamin and alpha-actinin during all stages of muscle development. By contrast, colocalization with its previously identified binding partner zyxin is restricted to early developmental stages. Genetic and cellular analyses of skeletal muscle provided direct evidence for an alternative transcriptional start site in exon three, corroborating the expression of a myopodin variant lacking the PDZ domain encoded by exons 1 and 2 in skeletal muscle. We conclude that myopodin is a multiadapter protein of the sarcomeric Z-disc that links nascent myofibrils to the sarcolemma via zyxin, and might play a role in early assembly and stabilization of the Z-disc. Mutations in FLNC, ACTN2 and several other genes encoding Z-disc-related proteins cause myopathy and cardiomyopathy. Its localization and its association with the myopathy-associated proteins filamin C and alpha-actinin make myopodin an interesting candidate for a muscle disease gene.
Collapse
Affiliation(s)
- Anja Linnemann
- Institute for Cell Biology, Department of Molecular Cell Biology, University of Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Jin L, Hastings NE, Blackman BR, Somlyo AV. Mechanical properties of the extracellular matrix alter expression of smooth muscle protein LPP and its partner palladin; relationship to early atherosclerosis and vascular injury. J Muscle Res Cell Motil 2009; 30:41-55. [PMID: 19205907 DOI: 10.1007/s10974-009-9173-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Accepted: 01/08/2009] [Indexed: 12/12/2022]
Abstract
Lipoma preferred partner (LPP) localizes to focal adhesions/dense bodies, is selectively expressed in smooth muscle cells (SMC) and enhances cell migration. SMCs cultured on denatured collagen or on a rigid substrate, up regulated expression of LPP, its partner palladin, tenascin C (TN-C), phosphorylated focal adhesion kinase (pFAK) and exhibited robust stress fibers. In an endothelial (EC)/SMC hemodynamic flow system, shear stress waveforms mimicking atheroprone flow, applied to the EC layer, significantly decreased expression of SMC LPP and palladin. They were also down regulated with TN-C, in an ApoE murine model of atherosclerosis and with oxidative stress but up regulated in an arterial injury model in response to upstream sequential changes in pFAK, Prx1 and TN-C. In conclusion, expression of LPP and palladin are modulated by a mix of mechanical cues, oxidative stress and substrate composition which translate into their up or down regulation in vessel wall injury and early atherogenesis.
Collapse
Affiliation(s)
- Li Jin
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
9
|
Morrow D, Guha S, Sweeney C, Birney Y, Walshe T, O’Brien C, Walls D, Redmond EM, Cahill PA. Notch and Vascular Smooth Muscle Cell Phenotype. Circ Res 2008; 103:1370-82. [PMID: 19059839 DOI: 10.1161/circresaha.108.187534] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Notch signaling pathway is critical for cell fate determination during embryonic development, including many aspects of vascular development. An emerging paradigm suggests that the Notch gene regulatory network is often recapitulated in the context of phenotypic modulation of vascular smooth muscle cells (VSMC), vascular remodeling, and repair in adult vascular disease following injury. Notch ligand receptor interactions lead to cleavage of receptor, translocation of the intracellular receptor (Notch IC), activation of transcriptional CBF-1/RBP-Jκ–dependent and –independent pathways, and transduction of downstream Notch target gene expression. Hereditary mutations of Notch components are associated with congenital defects of the cardiovascular system in humans such as Alagille syndrome and cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Recent loss- or gain-of-function studies have provided insight into novel Notch-mediated CBF-1/RBP-Jκ–dependent and –independent signaling and cross-regulation to other molecules that may play a critical role in VSMC phenotypic switching. Notch receptors are critical for controlling VSMC differentiation and dictating the phenotypic response following vascular injury through interaction with a triad of transcription factors that act synergistically to regulate VSMC differentiation. This review focuses on the role of Notch receptor ligand interactions in dictating VSMC behavior and phenotype and presents recent findings on the molecular interactions between the Notch components and VSMC-specific genes to further understand the function of Notch signaling in vascular tissue and disease.
Collapse
Affiliation(s)
- David Morrow
- From the Vascular Health Research Centre (D.M., S.G., C.S., Y.B., T.W., P.A.C.), Faculty of Science and Health; and School of Biotechnology (D.W.), National Centre for Sensor Research, Dublin City University, Ireland; Department of Surgery (D.M., E.M.R.), University of Rochester, NY; Schepens Eye Research Institute (T.W.), Harvard Medical School, Boston, Mass; and Mater Misericordiae Hospital (C.O.), Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin,
| | - Shaunta Guha
- From the Vascular Health Research Centre (D.M., S.G., C.S., Y.B., T.W., P.A.C.), Faculty of Science and Health; and School of Biotechnology (D.W.), National Centre for Sensor Research, Dublin City University, Ireland; Department of Surgery (D.M., E.M.R.), University of Rochester, NY; Schepens Eye Research Institute (T.W.), Harvard Medical School, Boston, Mass; and Mater Misericordiae Hospital (C.O.), Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin,
| | - Catherine Sweeney
- From the Vascular Health Research Centre (D.M., S.G., C.S., Y.B., T.W., P.A.C.), Faculty of Science and Health; and School of Biotechnology (D.W.), National Centre for Sensor Research, Dublin City University, Ireland; Department of Surgery (D.M., E.M.R.), University of Rochester, NY; Schepens Eye Research Institute (T.W.), Harvard Medical School, Boston, Mass; and Mater Misericordiae Hospital (C.O.), Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin,
| | - Yvonne Birney
- From the Vascular Health Research Centre (D.M., S.G., C.S., Y.B., T.W., P.A.C.), Faculty of Science and Health; and School of Biotechnology (D.W.), National Centre for Sensor Research, Dublin City University, Ireland; Department of Surgery (D.M., E.M.R.), University of Rochester, NY; Schepens Eye Research Institute (T.W.), Harvard Medical School, Boston, Mass; and Mater Misericordiae Hospital (C.O.), Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin,
| | - Tony Walshe
- From the Vascular Health Research Centre (D.M., S.G., C.S., Y.B., T.W., P.A.C.), Faculty of Science and Health; and School of Biotechnology (D.W.), National Centre for Sensor Research, Dublin City University, Ireland; Department of Surgery (D.M., E.M.R.), University of Rochester, NY; Schepens Eye Research Institute (T.W.), Harvard Medical School, Boston, Mass; and Mater Misericordiae Hospital (C.O.), Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin,
| | - Colm O’Brien
- From the Vascular Health Research Centre (D.M., S.G., C.S., Y.B., T.W., P.A.C.), Faculty of Science and Health; and School of Biotechnology (D.W.), National Centre for Sensor Research, Dublin City University, Ireland; Department of Surgery (D.M., E.M.R.), University of Rochester, NY; Schepens Eye Research Institute (T.W.), Harvard Medical School, Boston, Mass; and Mater Misericordiae Hospital (C.O.), Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin,
| | - Dermot Walls
- From the Vascular Health Research Centre (D.M., S.G., C.S., Y.B., T.W., P.A.C.), Faculty of Science and Health; and School of Biotechnology (D.W.), National Centre for Sensor Research, Dublin City University, Ireland; Department of Surgery (D.M., E.M.R.), University of Rochester, NY; Schepens Eye Research Institute (T.W.), Harvard Medical School, Boston, Mass; and Mater Misericordiae Hospital (C.O.), Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin,
| | - Eileen M. Redmond
- From the Vascular Health Research Centre (D.M., S.G., C.S., Y.B., T.W., P.A.C.), Faculty of Science and Health; and School of Biotechnology (D.W.), National Centre for Sensor Research, Dublin City University, Ireland; Department of Surgery (D.M., E.M.R.), University of Rochester, NY; Schepens Eye Research Institute (T.W.), Harvard Medical School, Boston, Mass; and Mater Misericordiae Hospital (C.O.), Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin,
| | - Paul A. Cahill
- From the Vascular Health Research Centre (D.M., S.G., C.S., Y.B., T.W., P.A.C.), Faculty of Science and Health; and School of Biotechnology (D.W.), National Centre for Sensor Research, Dublin City University, Ireland; Department of Surgery (D.M., E.M.R.), University of Rochester, NY; Schepens Eye Research Institute (T.W.), Harvard Medical School, Boston, Mass; and Mater Misericordiae Hospital (C.O.), Institute of Ophthalmology, The Conway Institute of Biomolecular and Biomedical Research, Dublin,
| |
Collapse
|
10
|
Petit MM, Lindskog H, Larsson E, Wasteson P, Athley E, Breuer S, Angstenberger M, Hertfelder D, Mattsson E, Nordheim A, Nelander S, Lindahl P. Smooth Muscle Expression of Lipoma Preferred Partner Is Mediated by an Alternative Intronic Promoter That Is Regulated by Serum Response Factor/Myocardin. Circ Res 2008; 103:61-9. [DOI: 10.1161/circresaha.108.177436] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipoma preferred partner (LPP) was recently recognized as a smooth muscle marker that plays a role in smooth muscle cell migration. In this report, we focus on the transcriptional regulation of the LPP gene. In particular, we investigate whether LPP is directly regulated by serum response factor (SRF). We show that the LPP gene contains 3 evolutionarily conserved CArG boxes and that 1 of these is part of an alternative promoter in intron 2. Quantitative RT-PCR shows that this alternative promoter directs transcription specifically to smooth muscle containing tissues in vivo. By using chromatin immunoprecipitation, we demonstrate that 2 of the CArG boxes, including the promoter-associated CArG box, bind to endogenous SRF in cultured aortic smooth muscle cells. Electrophoretic mobility-shift assays show that the conserved CArG boxes bind SRF in vitro. In reporter experiments, we show that the alternative promoter has transcriptional capacity that is dependent on SRF/myocardin and that the promoter associated CArG box is required for that activity. Finally, we show by quantitative RT-PCR that the alternative promoter is strongly downregulated in SRF-deficient embryonic stem cells and in smooth muscle tissues derived from conditional SRF knockout mice. Collectively, our data demonstrate that expression of LPP in smooth muscle is mediated by an alternative promoter that is regulated by SRF/myocardin.
Collapse
Affiliation(s)
- Marleen M.R. Petit
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Henrik Lindskog
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Erik Larsson
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Per Wasteson
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Elisabeth Athley
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Silke Breuer
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Meike Angstenberger
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - David Hertfelder
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Erney Mattsson
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Alfred Nordheim
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Sven Nelander
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| | - Per Lindahl
- From the Wallenberg Laboratory (M.M.R.P., H.L., E.L., P.W., E.A., S.B., E.M., S.N., P.L.), Sahlgrenska University Hospital, Göteborg, Sweden; Institute of Biomedicine (E.L., P.W., P.L.), Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, University of Göteborg, Göteborg, Sweden; and Interfaculty Institute for Cell Biology (M.A., D.H., A.N.), Tuebingen University, Germany. Present address for M.M.R.P.: Department of Human Genetics, University of Leuven, Belgium. Present
| |
Collapse
|
11
|
Do two mutually exclusive gene modules define the phenotypic diversity of mammalian smooth muscle? Mol Genet Genomics 2008; 280:127-37. [PMID: 18509681 DOI: 10.1007/s00438-008-0349-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2007] [Accepted: 05/01/2008] [Indexed: 10/22/2022]
Abstract
Smooth muscle cells (SMCs) are key components of all hollow organs, where they perform contractile, synthetic and other functions. Unlike other muscle cells, SMCs are not terminally differentiated, but exhibit considerable phenotypic variation. Such variation is manifested both across disease states such as asthma and atherosclerosis, and physiological states such as pregnancy and wound healing. While there has been considerable investigation into the diversity of SMCs at the level of morphology and individual biomarkers, less is known about the diversity of SMCs at the level of the transcriptome. To explore this question, we performed an extensive statistical analysis that integrates 200 transcriptional profiles obtained in different SMC phenotypes and reference tissues. Our results point towards a non-trivial hypothesis: that transcriptional variation in different SMC phenotypes is characterized by coordinated differential expression of two mutually exclusive (anti-correlating) gene modules. The first of these modules (C) encodes 19 co-transcribed cell cycle associated genes, whereas the other module (E) encodes 41 co-transcribed extra-cellular matrix components. We propose that the positioning of smooth muscle cells along the C/E axis constitutes an important determinant of SMC phenotypes. In conclusion, our study introduces a new approach to assess phenotypic variation in smooth muscle cells, and is relevant as an example of how integrative bioinformatics analysis can shed light on not only terminal differentiated states but also subtler details in phenotypic variability. It also raises the broader question whether coordinated expression of gene modules is a common mechanism underlying phenotypic variability in mammalian cells.
Collapse
|
12
|
Van Loo P, Aerts S, Thienpont B, De Moor B, Moreau Y, Marynen P. ModuleMiner - improved computational detection of cis-regulatory modules: are there different modes of gene regulation in embryonic development and adult tissues? Genome Biol 2008; 9:R66. [PMID: 18394174 PMCID: PMC2643937 DOI: 10.1186/gb-2008-9-4-r66] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2007] [Revised: 03/07/2008] [Accepted: 04/07/2008] [Indexed: 12/23/2022] Open
Abstract
ModuleMiner detects cis-regulatory modules in a set of co-expressed genes in tissue-specific microarray clusters and in embryonic development datasets. We present ModuleMiner, a novel algorithm for computationally detecting cis-regulatory modules (CRMs) in a set of co-expressed genes. ModuleMiner outperforms other methods for CRM detection on benchmark data, and successfully detects CRMs in tissue-specific microarray clusters and in embryonic development gene sets. Interestingly, CRM predictions for differentiated tissues exhibit strong enrichment close to the transcription start site, whereas CRM predictions for embryonic development gene sets are depleted in this region.
Collapse
Affiliation(s)
- Peter Van Loo
- Department of Molecular and Developmental Genetics, VIB, Herestraat 49, B-3000 Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|
13
|
Volger OL, Fledderus JO, Kisters N, Fontijn RD, Moerland PD, Kuiper J, van Berkel TJ, Bijnens APJJ, Daemen MJAP, Pannekoek H, Horrevoets AJG. Distinctive expression of chemokines and transforming growth factor-beta signaling in human arterial endothelium during atherosclerosis. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:326-37. [PMID: 17591977 PMCID: PMC1941599 DOI: 10.2353/ajpath.2007.061196] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Knowledge about the in vivo role of endothelium in chronic human atherosclerosis has mostly been derived by insights from mouse models. Therefore, we set out to establish by microarray analyses the gene expression profiles of endothelium from human large arteries, as isolated by laser microbeam microdissection, having focal atherosclerosis of the early or the advanced stage. Within individual arteries, the endothelial transcriptomes of the lesional and unaffected sides were compared pairwise, thus limiting genetic and environmental confounders. Specific endothelial signature gene sets were identified with changed expression levels in either early (n = 718) or advanced atherosclerosis (n = 403), relative to their paired plaque-free controls. Gene set enrichment analysis identified distinct sets of chemokines and differential enrichments of nuclear factor-kappaB-, p53-, and transforming growth factor-beta-related genes in advanced plaques. Immunohistochemistry validated the discriminative value of corresponding endothelial protein expression between early (fractalkine/CX3CL1, IP10/CCL10, TBX18) or advanced (BAX, NFKB2) stages of atherosclerosis and versus their plaque-free controls. The functional involvement of transforming growth factor-beta signaling in directing its downstream gene repertoire was substantiated by a consistent detection of activated SMAD2 in advanced lesions. Thus, we identified truly common, local molecular denominators of pathological changes to vascular endothelium, with a marked distinction of endothelial phenotype between early and advanced plaques.
Collapse
Affiliation(s)
- Oscar L Volger
- Department of Medical Biochemistry, Room K1-114, Academic Medical Center, University of Amsterdam, Meibergdreef 15, NL-1105 AZ, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Jin L, Kern MJ, Otey CA, Wamhoff BR, Somlyo AV. Angiotensin II, focal adhesion kinase, and PRX1 enhance smooth muscle expression of lipoma preferred partner and its newly identified binding partner palladin to promote cell migration. Circ Res 2007; 100:817-25. [PMID: 17322171 DOI: 10.1161/01.res.0000261351.54147.de] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipoma preferred partner (LPP) is a proline rich LIM domain family protein highly expressed at plasma membrane dense bodies and focal adhesions in smooth muscle cells.(1) Using the C-terminus of LPP as bait in a yeast two hybrid system, palladin, an actin-associated protein was identified. The palladin interacting region of LPP was mapped to the first and second LIM domains. The N-terminus of palladin interacted with LPP both in vitro and in vivo, but not solely through its FPLPPP and FPPPP motifs. Like LPP, palladin, is highly expressed in differentiated smooth muscle, colocalized at focal adhesions, at isolated lamellipodia and at dense bodies in smooth muscle tissue. Both LPP and palladin enhanced cell migration and spreading. LPP and palladin expression was markedly decreased, in contrast to vinculin or paxillin, in migration defective focal adhesion kinase null cells, but was restored by expression of the paired-related homeobox gene-1 protein. We have previously shown in focal adhesion kinase null cells, that tetracycline induced expression of focal adhesion kinase upregulated expression of LPP(2) and now show upregulation of palladin, and paired-related homeobox gene-1 protein. The expression of both LPP and palladin, like smooth muscle alpha-actin, was increased by angiotensin II, regulated by actin dynamics, upregulated by myocardin and expressed in the neointima of injured aorta. Overall, the data suggest that the function of LPP and palladin is context dependent, that they play a critical role in cytoskeletal remodeling, respond to signals induced by vascular injury as well as signals that induce smooth muscle cell hypertrophy, such as angiotension II.
Collapse
Affiliation(s)
- Li Jin
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908-0736, USA
| | | | | | | | | |
Collapse
|
15
|
Gorenne I, Jin L, Yoshida T, Sanders JM, Sarembock IJ, Owens GK, Somlyo AP, Somlyo AV. LPP expression during in vitro smooth muscle differentiation and stent-induced vascular injury. Circ Res 2006; 98:378-85. [PMID: 16397143 DOI: 10.1161/01.res.0000202802.34727.fd] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lipoma preferred partner (LPP) has been identified as a protein highly expressed in smooth muscle (SM) tissues. The aim of the present study was to determine mechanisms that regulate LPP expression in an in vitro model of SM cell (SMC) differentiation and in stent-induced pig coronary vessel injury. All trans-retinoic acid treatment of A404 cells induced a strong increase in LPP, as well as SM alpha-actin, SM myosin heavy chain, and smoothelin mRNA levels, in a Rho kinase (ROK)-dependent manner. Adenovirus mediated overexpression of myocardin in A404 cells significantly increased LPP mRNA expression. Interestingly, inactivation of RhoA with C3-exoenzyme or treatment with ROK inhibitors strongly inhibited myocardin mRNA expression in retinoic acid-treated A404 cells or human iliac vein SMCs. LPP silencing with short interfering RNA significantly decreased SMC migration. LPP expression was also markedly decreased in focal adhesion kinase (FAK)-null cells known to have impaired migration but rescued with inducible expression of FAK. LPP expression in FAK-null fibroblasts enhanced cell spreading. In stented pig coronary vessels, LPP was expressed in the neointima of cells lacking smoothelin and showed expression patterns identical to those of SM alpha-actin. In conclusion, LPP appears to be a myocardin-, RhoA/ROK-dependent SMC differentiation marker that plays a role in regulating SMC migration.
Collapse
Affiliation(s)
- I Gorenne
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
Interactions between endothelial cells and mural cells (pericytes and vascular smooth muscle cells) in the blood vessel wall have recently come into focus as central processes in the regulation of vascular formation, stabilization, remodeling, and function. Failure of the interactions between the 2 cell types, as seen in numerous genetic mouse models, results in severe and often lethal cardiovascular defects. Abnormal interactions between the 2 cell types are also implicated in a number of human pathological conditions, including tumor angiogenesis, diabetic microangiopathy, ectopic tissue calcification, and stroke and dementia syndrome CADASIL. In the present review, we summarize current knowledge concerning the identity, characteristics, diversity, ontogeny, and plasticity of pericytes. We focus on the advancement in recent years of the understanding of intercellular communication between endothelial and mural cells with a focus on transforming growth factor beta, angiopoietins, platelet-derived growth factor, spingosine-1-phosphate, and Notch ligands and their respective receptors. We finally highlight recent important data contributing to the understanding of the role of pericytes in tumor angiogenesis, diabetic retinopathy, and hereditary lymphedema.
Collapse
Affiliation(s)
- Annika Armulik
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
17
|
Crombez KRMO, Vanoirbeek EMR, Van de Ven WJM, Petit MMR. Transactivation functions of the tumor-specific HMGA2/LPP fusion protein are augmented by wild-type HMGA2. Mol Cancer Res 2005; 3:63-70. [PMID: 15755872 DOI: 10.1158/1541-7786.mcr-04-0181] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The gene encoding the architectural transcription factor HMGA2 is frequently rearranged in several benign tumors of mesenchymal origin. The lipoma preferred partner (LPP) gene is the most frequent translocation partner of HMGA2 in a subgroup of lipomas, which are benign tumors of adipose tissue. In these lipomas, HMGA2/LPP fusion transcripts are expressed, which encode for the three AT-hooks of HMGA2 followed by the two most carboxyl-terminal LIM domains (protein-protein interaction domains) of LPP. Identical fusion transcripts are also expressed in other benign mesenchymal tumors. Previous studies revealed that the LIM domains of LPP have transcriptional activation capacity in GAL4-based luciferase reporter assays. Here, we show that the HMGA2/LPP fusion protein retains the transactivation functions of the LPP LIM domains and thus functions as transcription factor. The HMGA2/LPP fusion protein activates transcription from the well-characterized PRDII element, which is a part of the IFN-beta enhancer and which is known to bind to HMGA2. We also show that HMGA2/LPP activates transcription from the BAT-1 element of the rhodopsin promoter, a HMGA1-binding element. HMGA1 is a closely related family member of HMGA2. Finally, in a number of lipomas, HMGA2/LPP and HMGA2 are coexpressed, and HMGA2 augments the transactivation functions of HMGA2/LPP. These results support the concept that the transactivation functions of the novel HMGA2/LPP transcription factor contribute to lipomagenesis.
Collapse
Affiliation(s)
- Koen R M O Crombez
- Department of Human Genetics, University of Leuven and Flanders Interuniversity Institute for Biotechnology, Herestraat 49 bus 602, B-3000 Leuven, Belgium
| | | | | | | |
Collapse
|
18
|
Nelander S, Larsson E, Kristiansson E, Månsson R, Nerman O, Sigvardsson M, Mostad P, Lindahl P. Predictive screening for regulators of conserved functional gene modules (gene batteries) in mammals. BMC Genomics 2005; 6:68. [PMID: 15882449 PMCID: PMC1134656 DOI: 10.1186/1471-2164-6-68] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2004] [Accepted: 05/09/2005] [Indexed: 01/08/2023] Open
Abstract
Background The expression of gene batteries, genomic units of functionally linked genes which are activated by similar sets of cis- and trans-acting regulators, has been proposed as a major determinant of cell specialization in metazoans. We developed a predictive procedure to screen the mouse and human genomes and transcriptomes for cases of gene-battery-like regulation. Results In a screen that covered ~40 per cent of all annotated protein-coding genes, we identified 21 co-expressed gene clusters with statistically supported sharing of cis-regulatory sequence elements. 66 predicted cases of over-represented transcription factor binding motifs were validated against the literature and fell into three categories: (i) previously described cases of gene battery-like regulation, (ii) previously unreported cases of gene battery-like regulation with some support in a limited number of genes, and (iii) predicted cases that currently lack experimental support. The novel predictions include for example Sox 17 and RFX transcription factor binding sites that were detected in ~10% of all testis specific genes, and HNF-1 and 4 binding sites that were detected in ~30% of all kidney specific genes respectively. The results are publicly available at . Conclusion 21 co-expressed gene clusters were enriched for a total of 66 shared cis-regulatory sequence elements. A majority of these predictions represent novel cases of potential co-regulation of functionally coupled proteins. Critical technical parameters were evaluated, and the results and the methods provide a valuable resource for future experimental design.
Collapse
Affiliation(s)
- Sven Nelander
- Sahlgrenska Academy, Department of medical and physiological biochemistry Box 440, SE-405 30 Göteborg, Sweden
| | - Erik Larsson
- Sahlgrenska Academy, Department of medical and physiological biochemistry Box 440, SE-405 30 Göteborg, Sweden
| | - Erik Kristiansson
- Chalmers Technical University, Department of mathematical statistics, Eklandagatan 76, SE-412 96 Göteborg, Sweden
| | - Robert Månsson
- Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, BMC B10, Klinikgatan 26, SE-221 48 Lund, Sweden
| | - Olle Nerman
- Chalmers Technical University, Department of mathematical statistics, Eklandagatan 76, SE-412 96 Göteborg, Sweden
| | - Mikael Sigvardsson
- Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, BMC B10, Klinikgatan 26, SE-221 48 Lund, Sweden
| | - Petter Mostad
- Chalmers Technical University, Department of mathematical statistics, Eklandagatan 76, SE-412 96 Göteborg, Sweden
| | - Per Lindahl
- Sahlgrenska Academy, Department of medical and physiological biochemistry Box 440, SE-405 30 Göteborg, Sweden
| |
Collapse
|
19
|
Yamashita R, Suzuki Y, Sugano S, Nakai K. Genome-wide analysis reveals strong correlation between CpG islands with nearby transcription start sites of genes and their tissue specificity. Gene 2005; 350:129-36. [PMID: 15784181 DOI: 10.1016/j.gene.2005.01.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2004] [Revised: 12/28/2004] [Accepted: 01/24/2005] [Indexed: 10/25/2022]
Abstract
It has been envisaged that CpG islands are often observed near the transcriptional start sites (TSS) of housekeeping genes. However, neither the precise positions of CpG islands relative to TSS of genes nor the correlation between the presence of the CpG islands and the expression specificity of these genes is well-understood. Using thousands of sequences with known TSS in human and mouse, we found that there is a clear peak in the distribution of CpG islands around TSS in the genes of these two species. Thus, we classified human (mouse) genes into 6600 (2948) CpG+ genes and 2619 (1830) CpG- ones, based on the presence of a CpG island within the -100: +100 region. We estimated the degree of each gene being a housekeeper by the number of cDNA libraries where its ESTs were detected. Then, the tendency that a gene lacking CpG islands around its TSS is expressed with a higher degree of tissue specificity turned out to be evolutionarily conserved. We also confirmed this tendency by analyzing the gene ontology annotation of classified genes. Since no such clear correlation was found in the control data (mRNAs, pre-mRNAs, and chromosome banding pattern), we concluded that the effect of a CpG island near the TSS should be more important than the global GC content of the region where the gene resides.
Collapse
Affiliation(s)
- Riu Yamashita
- Human Genome Center, Institute of Medical Science, University of Tokyo, 4-6-1, Shirokane-dai Minato-ku, Tokyo 108-8639, Japan
| | | | | | | |
Collapse
|
20
|
The tumor suppressor Scrib interacts with the zyxin-related protein LPP, which shuttles between cell adhesion sites and the nucleus. BMC Cell Biol 2005; 6:1. [PMID: 15649318 PMCID: PMC546208 DOI: 10.1186/1471-2121-6-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2004] [Accepted: 01/13/2005] [Indexed: 11/22/2022] Open
Abstract
Background At sites of cell adhesion, proteins exist that not only perform structural tasks but also have a signaling function. Previously, we found that the Lipoma Preferred Partner (LPP) protein is localized at sites of cell adhesion such as focal adhesions and cell-cell contacts, and shuttles to the nucleus where it has transcriptional activation capacity. LPP is a member of the zyxin family of proteins, which contains five members: ajuba, LIMD1, LPP, TRIP6 and zyxin. LPP has three LIM domains (zinc-finger protein interaction domains) at its carboxy-terminus, which are preceded by a proline-rich pre-LIM region containing a number of protein interaction domains. Results To catch the role of LPP at sites of cell adhesion, we made an effort to identify binding partners of LPP. We found the tumor suppressor protein Scrib, which is a component of cell-cell contacts, as interaction partner of LPP. Human Scrib, which is a functional homologue of Drosophila scribble, is a member of the leucine-rich repeat and PDZ (LAP) family of proteins that is involved in the regulation of cell adhesion, cell shape and polarity. In addition, Scrib displays tumor suppressor activity. The binding between Scrib and LPP is mediated by the PDZ domains of Scrib and the carboxy-terminus of LPP. Both proteins localize in cell-cell contacts. Whereas LPP is also localized in focal adhesions and in the nucleus, Scrib could not be detected at these locations in MDCKII and CV-1 cells. Furthermore, our investigations indicate that Scrib is dispensable for targeting LPP to focal adhesions and to cell-cell contacts, and that LPP is not necessary for localizing Scrib in cell-cell contacts. We show that all four PDZ domains of Scrib are dispensable for localizing this protein in cell-cell contacts. Conclusions Here, we identified an interaction between one of zyxin's family members, LPP, and the tumor suppressor protein Scrib. Both proteins localize in cell-cell contacts. This interaction links Scrib to a communication pathway between cell-cell contacts and the nucleus, and implicates LPP in Scrib-associated functions.
Collapse
|