51
|
What do polymorphisms tell us about the mechanisms of COPD? Clin Sci (Lond) 2017; 131:2847-2863. [PMID: 29203722 DOI: 10.1042/cs20160718] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 10/22/2017] [Accepted: 11/01/2017] [Indexed: 12/11/2022]
Abstract
COPD (chronic obstructive pulmonary disease) is characterized by irreversible lung airflow obstruction. Cigarette smoke is the major risk factor for COPD development. However, only a minority number of smokers develop COPD, and there are substantial variations in lung function among smokers, suggesting that genetic determinants in COPD susceptibility. During the past decade, genome-wide association studies and exome sequencing have been instrumental to identify the genetic determinants of complex traits, including COPD. Focused studies have revealed mechanisms by which genetic variants contribute to COPD and have led to novel insights in COPD pathogenesis. Through functional investigations of causal variants in COPD, from the proteinase-antiproteinase theory to emerging roles of developmental pathways (such as Hedgehog and Wnt pathways) in COPD, we have greatly expanded our understanding on this complex pulmonary disease. In this review, we critically review functional investigations on roles of genetic polymorphisms in COPD, and discuss future challenges and opportunities in discovering novel mechanisms of functional variants.
Collapse
|
52
|
Kardon G, Ackerman KG, McCulley DJ, Shen Y, Wynn J, Shang L, Bogenschutz E, Sun X, Chung WK. Congenital diaphragmatic hernias: from genes to mechanisms to therapies. Dis Model Mech 2017; 10:955-970. [PMID: 28768736 PMCID: PMC5560060 DOI: 10.1242/dmm.028365] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Congenital diaphragmatic hernias (CDHs) and structural anomalies of the diaphragm are a common class of congenital birth defects that are associated with significant morbidity and mortality due to associated pulmonary hypoplasia, pulmonary hypertension and heart failure. In ∼30% of CDH patients, genomic analyses have identified a range of genetic defects, including chromosomal anomalies, copy number variants and sequence variants. The affected genes identified in CDH patients include transcription factors, such as GATA4, ZFPM2, NR2F2 and WT1, and signaling pathway components, including members of the retinoic acid pathway. Mutations in these genes affect diaphragm development and can have pleiotropic effects on pulmonary and cardiac development. New therapies, including fetal endoscopic tracheal occlusion and prenatal transplacental fetal treatments, aim to normalize lung development and pulmonary vascular tone to prevent and treat lung hypoplasia and pulmonary hypertension, respectively. Studies of the association between particular genetic mutations and clinical outcomes should allow us to better understand the origin of this birth defect and to improve our ability to predict and identify patients most likely to benefit from specialized treatment strategies.
Collapse
Affiliation(s)
- Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Kate G Ackerman
- Departments of Pediatrics (Critical Care) and Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David J McCulley
- Department of Pediatrics, University of Wisconsin, Madison, WI 53792, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Julia Wynn
- Departments of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Linshan Shang
- Departments of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
| | - Eric Bogenschutz
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wendy K Chung
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
53
|
Bultmann-Mellin I, Dinger K, Debuschewitz C, Loewe KMA, Melcher Y, Plum MTW, Appel S, Rappl G, Willenborg S, Schauss AC, Jüngst C, Krüger M, Dressler S, Nakamura T, Wempe F, Alejandre Alcázar MA, Sterner-Kock A. Role of LTBP4 in alveolarization, angiogenesis, and fibrosis in lungs. Am J Physiol Lung Cell Mol Physiol 2017; 313:L687-L698. [PMID: 28684544 DOI: 10.1152/ajplung.00031.2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/22/2017] [Accepted: 06/29/2017] [Indexed: 12/31/2022] Open
Abstract
Deficiency of the extracellular matrix protein latent transforming growth factor-β (TGF-β)-binding protein-4 (LTBP4) results in lack of intact elastic fibers, which leads to disturbed pulmonary development and lack of normal alveolarization in humans and mice. Formation of alveoli and alveolar septation in pulmonary development requires the concerted interaction of extracellular matrix proteins, growth factors such as TGF-β, fibroblasts, and myofibroblasts to promote elastogenesis as well as vascular formation in the alveolar septae. To investigate the role of LTBP4 in this context, lungs of LTBP4-deficient (Ltbp4-/-) mice were analyzed in close detail. We elucidate the role of LTBP4 in pulmonary alveolarization and show that three different, interacting mechanisms might contribute to alveolar septation defects in Ltbp4-/- lungs: 1) absence of an intact elastic fiber network, 2) reduced angiogenesis, and 3) upregulation of TGF-β activity resulting in profibrotic processes in the lung.
Collapse
Affiliation(s)
- Insa Bultmann-Mellin
- Center for Experimental Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Katharina Dinger
- Center for Experimental Medicine, Medical Faculty, University of Cologne, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Carolin Debuschewitz
- Center for Experimental Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Katharina M A Loewe
- Center for Experimental Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Yvonne Melcher
- Center for Experimental Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Miro T W Plum
- Center for Experimental Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Sarah Appel
- Department of Pediatrics and Adolescent Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Gunter Rappl
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany
| | | | - Astrid C Schauss
- Cluster of Excellence, Cellular Stress Response in Aging-Related Diseases, Core Facility Imaging, University of Cologne, Cologne, Germany
| | - Christian Jüngst
- Cluster of Excellence, Cellular Stress Response in Aging-Related Diseases, Core Facility Imaging, University of Cologne, Cologne, Germany
| | - Marcus Krüger
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cluster of Excellence, Cellular Stress Response in Aging-Related Diseases, Core Facility Proteomics, University of Cologne, Cologne, Germany.,Institute for Genetics, University of Cologne, Cologne, Germany
| | - Sven Dressler
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Tomoyuki Nakamura
- Department of Pharmacology, Kansai Medical University, Osaka, Japan; and
| | - Frank Wempe
- Department of Molecular Hematology, University of Frankfurt Medical School, Frankfurt am Main, Germany
| | - Miguel A Alejandre Alcázar
- Department of Pediatrics and Adolescent Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | - Anja Sterner-Kock
- Center for Experimental Medicine, Medical Faculty, University of Cologne, Cologne, Germany;
| |
Collapse
|
54
|
Fujikawa Y, Yoshida H, Inoue T, Ohbayashi T, Noda K, von Melchner H, Iwasaka T, Shiojima I, Akama TO, Nakamura T. Latent TGF-β binding protein 2 and 4 have essential overlapping functions in microfibril development. Sci Rep 2017; 7:43714. [PMID: 28252045 PMCID: PMC5333096 DOI: 10.1038/srep43714] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/27/2017] [Indexed: 11/09/2022] Open
Abstract
Microfibrils are exracellular matrix components necessary for elastic fiber assembly and for suspending lenses. We previously reported that latent TGF-β binding protein 2 (LTBP-2), a microfibril-associated protein, is required for forming stable microfibril bundles in ciliary zonules. However, it was not understood why Ltbp2 null mice only showed an eye-specific phenotype, whereas LTBP-2 is abundantly expressed in other tissues containing microfibrils in wild type mice. Here, we show that LTBP-4, another microfibril-associated protein, compensates for the loss of LTBP-2 in microfibril formation. Ltbp2/4S double knockout (DKO) mice showed increased lethality due to emphysema, which was much more severe than that found in Ltbp4S null mice. Elastic fibers in the lungs of Ltbp2/4S DKO mice were severely disorganized and fragmented. Cultured mouse embryonic fibroblasts (MEFs) from Ltbp2/4S DKO embryos developed reduced microfibril meshwork in serum-free conditions, whereas the microfibril formation was restored by the addition of either recombinant LTBP-2 or -4. Finally, ectopic expression of LTBP-4 in the whole body restored ciliary zonule microfibril bundles in the eyes of Ltbp2 null mice. These data suggest that LTBP-2 and -4 have critical overlapping functions in forming the robust structure of microfibrils in vitro and in vivo.
Collapse
Affiliation(s)
- Yusuke Fujikawa
- Department of Pharmacology, Kansai Medical University, Osaka, 573-1010, Japan.,Department of Cardiology, Kansai Medical University, Osaka, 573-1010, Japan
| | - Hideyuki Yoshida
- Department of Pharmacology, Kansai Medical University, Osaka, 573-1010, Japan.,Department of Ophthalmology, Kansai Medical University, Osaka, 753-1010, Japan
| | - Tadashi Inoue
- Department of Pharmacology, Kansai Medical University, Osaka, 573-1010, Japan.,Department of Plastic and Reconstructive Surgery, Kansai Medical University, Osaka, 573-1010, Japan
| | - Tetsuya Ohbayashi
- Division of Laboratory Animal Science, Research Center for Bioscience and Technology, Tottori University Graduate School of Medical Sciences, Yonago, Tottori, 683-8503, Japan
| | - Kazuo Noda
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507, Japan
| | - Harald von Melchner
- Department of Molecular Hematology, University of Frankfurt Medical School, Frankfurt am Main, 60590, Germany
| | - Toshiji Iwasaka
- Department of Cardiology, Kansai Medical University, Osaka, 573-1010, Japan
| | - Ichiro Shiojima
- Department of Cardiology, Kansai Medical University, Osaka, 573-1010, Japan
| | - Tomoya O Akama
- Department of Pharmacology, Kansai Medical University, Osaka, 573-1010, Japan
| | - Tomoyuki Nakamura
- Department of Pharmacology, Kansai Medical University, Osaka, 573-1010, Japan
| |
Collapse
|
55
|
Gkogkolou P, Hildebrandt K, Broekaert S, Metze D, Sengle G, Böhm M. Cutis laxa acquisita: novel insights into impaired elastic fibre regeneration. Br J Dermatol 2017; 176:832-835. [DOI: 10.1111/bjd.15196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- P. Gkogkolou
- Department of Dermatology; University of Münster; Münster Germany
| | - K. Hildebrandt
- Center for Biochemistry; Medical Faculty; University of Cologne; Cologne Germany
- Center for Molecular Medicine Cologne (CMMC); University of Cologne; Cologne Germany
| | - S. Broekaert
- Department of Dermatology; University of Münster; Münster Germany
| | - D. Metze
- Department of Dermatology; University of Münster; Münster Germany
| | - G. Sengle
- Center for Biochemistry; Medical Faculty; University of Cologne; Cologne Germany
- Center for Molecular Medicine Cologne (CMMC); University of Cologne; Cologne Germany
| | - M. Böhm
- Department of Dermatology; University of Münster; Münster Germany
| |
Collapse
|
56
|
Determinants of orofacial clefting I: Effects of 5-Aza-2'-deoxycytidine on cellular processes and gene expression during development of the first branchial arch. Reprod Toxicol 2016; 67:85-99. [PMID: 27915011 DOI: 10.1016/j.reprotox.2016.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/19/2016] [Accepted: 11/29/2016] [Indexed: 12/27/2022]
Abstract
In this study, we identify gene targets and cellular events mediating the teratogenic action(s) of 5-Aza-2'-deoxycytidine (AzaD), an inhibitor of DNA methylation, on secondary palate development. Exposure of pregnant mice (on gestation day (GD) 9.5) to AzaD for 12h resulted in the complete penetrance of cleft palate (CP) in fetuses. Analysis of cells of the embryonic first branchial arch (1-BA), in fetuses exposed to AzaD, revealed: 1) significant alteration in expression of genes encoding several morphogenetic factors, cell cycle inhibitors and regulators of apoptosis; 2) a decrease in cell proliferation; and, 3) an increase in apoptosis. Pyrosequencing of selected genes, displaying pronounced differential expression in AzaD-exposed 1-BAs, failed to reveal significant alterations in CpG methylation levels in their putative promoters or gene bodies. CpG methylation analysis suggested that the effects of AzaD on gene expression were likely indirect.
Collapse
|
57
|
Cohen R, Halevy A, Aharoni S, Kraus D, Konen O, Basel-Vanagaite L, Goldberg-Stern H, Straussberg R. Polymicrogyria and myoclonic epilepsy in autosomal recessive cutis laxa type 2A. Neurogenetics 2016; 17:251-257. [PMID: 27631729 DOI: 10.1007/s10048-016-0491-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/17/2016] [Indexed: 11/27/2022]
Abstract
Cutis laxa syndromes are rare inherited disorders of skin and connective tissue metabolism associated with variable systemic involvement. The main clinical manifestation is loose, wrinkled, redundant, inelastic skin, hypotonia, typical facies including short nose and down-slanting palpebral fissures, and varying degrees of developmental delay. The aim of this report is to describe two siblings diagnosed with a moderate form of ATP6V0A2-related cutis laxa with polymicrogyria (cobblestone-like brain dysgenesis). One of the patients has myoclonic epilepsy which may have contributed to his more severe clinical presentation. The literature on cutis laxa syndromes is reviewed.
Collapse
Affiliation(s)
- Rony Cohen
- Department of Pediatric Neurology and Epilepsy Center, Schneider Children's Medical Center of Israel, Petach Tikva, 4920235, Israel. .,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Ayelet Halevy
- Department of Pediatric Neurology and Epilepsy Center, Schneider Children's Medical Center of Israel, Petach Tikva, 4920235, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Aharoni
- Department of Pediatric Neurology and Epilepsy Center, Schneider Children's Medical Center of Israel, Petach Tikva, 4920235, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dror Kraus
- Department of Pediatric Neurology and Epilepsy Center, Schneider Children's Medical Center of Israel, Petach Tikva, 4920235, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Konen
- Department of Radiology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lina Basel-Vanagaite
- Pediatric Genetic Service, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Raphael Recanati Genetics Institute, Rabin Medical Center - Beilinson Hospital, Petach Tikva, Israel
| | - Hadassa Goldberg-Stern
- Department of Pediatric Neurology and Epilepsy Center, Schneider Children's Medical Center of Israel, Petach Tikva, 4920235, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rachel Straussberg
- Department of Pediatric Neurology and Epilepsy Center, Schneider Children's Medical Center of Israel, Petach Tikva, 4920235, Israel.,Pediatric Genetic Service, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
58
|
Bultmann-Mellin I, Essers J, van Heijingen PM, von Melchner H, Sengle G, Sterner-Kock A. Function of Ltbp-4L and fibulin-4 in survival and elastogenesis in mice. Dis Model Mech 2016; 9:1367-1374. [PMID: 27585882 PMCID: PMC5117228 DOI: 10.1242/dmm.026005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/15/2016] [Indexed: 12/18/2022] Open
Abstract
LTBP-4L and LTBP-4S are two isoforms of the extracellular matrix protein latent-transforming growth factor beta-binding protein 4 (LTBP-4). The mutational inactivation of both isoforms causes autosomal recessive cutis laxa type 1C (ARCL1C) in humans and an ARCL1C-like phenotype in Ltbp4-/- mice, both characterized by high postnatal mortality and severely affected elastogenesis. However, genetic data in mice suggest isoform-specific functions for Ltbp-4 because Ltbp4S-/- mice, solely expressing Ltbp-4L, survive to adulthood. This clearly suggests a requirement of Ltbp-4L for postnatal survival. A major difference between Ltbp4S-/- and Ltbp4-/- mice is the matrix incorporation of fibulin-4 (a key factor for elastogenesis; encoded by the Efemp2 gene), which is normal in Ltbp4S-/- mice, whereas it is defective in Ltbp4-/- mice, suggesting that the presence of Ltbp-4L might be required for this process. To investigate the existence of a functional interaction between Ltbp-4L and fibulin-4, we studied the consequences of fibulin-4 deficiency in mice only expressing Ltbp-4L. Resulting Ltbp4S-/-;Fibulin-4R/R mice showed a dramatically reduced lifespan compared to Ltbp4S-/- or Fibulin-4R/R mice, which survive to adulthood. This dramatic reduction in survival of Ltbp4S-/-;Fibulin-4R/R mice correlates with severely impaired elastogenesis resulting in defective alveolar septation and distal airspace enlargement in lung, and increased aortic wall thickness with severely fragmented elastic lamellae. Additionally, Ltbp4S-/-;Fibulin-4R/R mice suffer from aortic aneurysm formation combined with aortic tortuosity, in contrast to Ltbp4S-/- or Fibulin-4R/R mice. Together, in accordance with our previous biochemical findings of a physical interaction between Ltbp-4L and fibulin-4, these novel in vivo data clearly establish a functional link between Ltbp-4L and fibulin-4 as a crucial molecular requirement for survival and elastogenesis in mice.
Collapse
Affiliation(s)
- Insa Bultmann-Mellin
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Jeroen Essers
- Department of Molecular Genetics, Cancer Genomics Centre, Erasmus MC, 3015 CN Rotterdam, The Netherlands.,Department of Radiation Oncology, Erasmus MC, 3015 CN Rotterdam, The Netherlands.,Department of Vascular Surgery, Erasmus MC, 3015 CN Rotterdam, The Netherlands
| | - Paula M van Heijingen
- Department of Molecular Genetics, Cancer Genomics Centre, Erasmus MC, 3015 CN Rotterdam, The Netherlands
| | - Harald von Melchner
- Department of Molecular Hematology, University of Frankfurt Medical School, 60590 Frankfurt am Main, Germany
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Anja Sterner-Kock
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
59
|
Robertson IB, Rifkin DB. Regulation of the Bioavailability of TGF-β and TGF-β-Related Proteins. Cold Spring Harb Perspect Biol 2016; 8:8/6/a021907. [PMID: 27252363 DOI: 10.1101/cshperspect.a021907] [Citation(s) in RCA: 290] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The bioavailability of members of the transforming growth factor β (TGF-β) family is controlled by a number of mechanisms. Bona fide TGF-β is sequestered into the matrix in a latent state and must be activated before it can bind to its receptors. Here, we review the molecules and mechanisms that regulate the bioavailability of TGF-β and compare these mechanisms with those used to regulate other TGF-β family members. We also assess the physiological significance of various latent TGF-β activators, as well as other extracellular modulators of TGF-β family signaling, by examining the available in vivo data from knockout mouse models and other biological systems.
Collapse
Affiliation(s)
- Ian B Robertson
- Departments of Cell Biology, New York University School of Medicine, New York, New York 10016
| | - Daniel B Rifkin
- Departments of Cell Biology, New York University School of Medicine, New York, New York 10016 Departments of Medicine, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
60
|
Abstract
The bioavailability of members of the transforming growth factor β (TGF-β) family is controlled by a number of mechanisms. Bona fide TGF-β is sequestered into the matrix in a latent state and must be activated before it can bind to its receptors. Here, we review the molecules and mechanisms that regulate the bioavailability of TGF-β and compare these mechanisms with those used to regulate other TGF-β family members. We also assess the physiological significance of various latent TGF-β activators, as well as other extracellular modulators of TGF-β family signaling, by examining the available in vivo data from knockout mouse models and other biological systems.
Collapse
Affiliation(s)
- Ian B Robertson
- Departments of Cell Biology, New York University School of Medicine, New York, New York 10016
| | - Daniel B Rifkin
- Departments of Cell Biology, New York University School of Medicine, New York, New York 10016 Departments of Medicine, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
61
|
Overexpression of Latent TGFβ Binding Protein 4 in Muscle Ameliorates Muscular Dystrophy through Myostatin and TGFβ. PLoS Genet 2016; 12:e1006019. [PMID: 27148972 PMCID: PMC4858180 DOI: 10.1371/journal.pgen.1006019] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 04/08/2016] [Indexed: 01/06/2023] Open
Abstract
Latent TGFβ binding proteins (LTBPs) regulate the extracellular availability of latent TGFβ. LTBP4 was identified as a genetic modifier of muscular dystrophy in mice and humans. An in-frame insertion polymorphism in the murine Ltbp4 gene associates with partial protection against muscular dystrophy. In humans, nonsynonymous single nucleotide polymorphisms in LTBP4 associate with prolonged ambulation in Duchenne muscular dystrophy. To better understand LTBP4 and its role in modifying muscular dystrophy, we created transgenic mice overexpressing the protective murine allele of LTBP4 specifically in mature myofibers using the human skeletal actin promoter. Overexpression of LTBP4 protein was associated with increased muscle mass and proportionally increased strength compared to age-matched controls. In order to assess the effects of LTBP4 in muscular dystrophy, LTBP4 overexpressing mice were bred to mdx mice, a model of Duchenne muscular dystrophy. In this model, increased LTBP4 led to greater muscle mass with proportionally increased strength, and decreased fibrosis. The increase in muscle mass and reduction in fibrosis were similar to what occurs when myostatin, a related TGFβ family member and negative regulator of muscle mass, was deleted in mdx mice. Supporting this, we found that myostatin forms a complex with LTBP4 and that overexpression of LTBP4 led to a decrease in myostatin levels. LTBP4 also interacted with TGFβ and GDF11, a protein highly related to myostatin. These data identify LTBP4 as a multi-TGFβ family ligand binding protein with the capacity to modify muscle disease through overexpression. Muscular dystrophy is a genetic disease with muscle weakness, replacement of muscle tissue with fibrosis, and premature death. The gene for latent TGFβ binding protein 4 (LTBP4) was previously found to modify muscular dystrophy in both mice and humans with variants that confer protection from disease. In order to better understand this modifier gene, the protective version of LTBP4 was overexpressed specifically in the skeletal muscles of mice. Increased levels of LTBP4 protein resulted in increased muscle mass. Overexpression of LTBP4 in a mouse model of Duchenne muscular dystrophy alleviated many disease-associated features producing larger muscles, increased strength, and reduced fibrosis in muscle. LTBP4 formed a complex with myostatin, a protein that when inhibited leads to muscle growth. In LTBP4-overexpressing mice, active myostatin protein was decreased. This study shows that LTBP4 modifies muscular dystrophy based on its ability to scaffold and regulate multiple TGFβ family members including myostatin.
Collapse
|
62
|
Vuckovic A, Herber-Jonat S, Flemmer AW, Ruehl IM, Votino C, Segers V, Benachi A, Martinovic J, Nowakowska D, Dzieniecka M, Jani JC. Increased TGF-β: a drawback of tracheal occlusion in human and experimental congenital diaphragmatic hernia? Am J Physiol Lung Cell Mol Physiol 2015; 310:L311-27. [PMID: 26637634 DOI: 10.1152/ajplung.00122.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 12/03/2015] [Indexed: 12/15/2022] Open
Abstract
Survivors of severe congenital diaphragmatic hernia (CDH) present significant respiratory morbidity despite lung growth induced by fetal tracheal occlusion (TO). We hypothesized that the underlying mechanisms would involve changes in lung extracellular matrix and dysregulated transforming growth factor (TGF)-β pathway, a key player in lung development and repair. Pulmonary expression of TGF-β signaling components, downstream effectors, and extracellular matrix targets were evaluated in CDH neonates who died between birth and the first few weeks of life after prenatal conservative management or TO, and in rabbit pups that were prenatally randomized for surgical CDH and TO vs. sham operation. Before tissue harvesting, lung tissue mechanics in rabbits was measured using the constant-phase model during the first 30 min of life. Human CDH and control fetal lungs were also collected from midterm onwards. Human and experimental CDH did not affect TGF-β/Smad2/3 expression and activity. In human and rabbit CDH lungs, TO upregulated TGF-β transcripts. Analysis of downstream pathways indicated increased Rho-associated kinases to the detriment of Smad2/3 activation. After TO, subtle accumulation of collagen and α-smooth muscle actin within alveolar walls was detected in rabbit pups and human CDH lungs with short-term mechanical ventilation. Despite TO-induced lung growth, mediocre lung tissue mechanics in the rabbit model was associated with increased transcription of extracellular matrix components. These results suggest that prenatal TO increases TGF-β/Rho kinase pathway, myofibroblast differentiation, and matrix deposition in neonatal rabbit and human CDH lungs. Whether this might influence postnatal development of sustainably ventilated lungs remains to be determined.
Collapse
Affiliation(s)
- Aline Vuckovic
- Laboratory of Physiology and Pathophysiology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium;
| | - Susanne Herber-Jonat
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Andreas W Flemmer
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Ina M Ruehl
- Division of Neonatology, University Children's Hospital, Perinatal Center, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Carmela Votino
- Department of Obstetrics and Gynecology, Fetal Medicine Unit, University Hospital Brugmann, Université Libre de Bruxelles, Brussels, Belgium
| | - Valérie Segers
- Unit of Pediatric Pathology, Pathology Department, University Hospital Brugmann, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexandra Benachi
- Department of Obstetrics and Gynecology and Centre de Maladie Rare: Hernie de Coupole Diaphragmatique, Hôpital Antoine Béclère, Assistance Publique Hôpitaux de Paris (APHP), Université Paris Sud, Paris, France
| | - Jelena Martinovic
- Unit of Fetal Pathology, Hôpital Antoine Béclère, Assistance Publique Hôpitaux de Paris (APHP), Université Paris Sud, Paris, France
| | - Dorota Nowakowska
- Department of Fetal-Maternal Medicine and Gynecology, Medical University and the Research Institute Polish Mother's Memorial Hospital, Lodz, Poland; and
| | - Monika Dzieniecka
- Department of Clinical Pathology, Medical University and the Research Institute Polish Mother's Memorial Hospital, Lodz, Poland
| | - Jacques C Jani
- Department of Obstetrics and Gynecology, Fetal Medicine Unit, University Hospital Brugmann, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
63
|
Fischer-Zirnsak B, Escande-Beillard N, Ganesh J, Tan Y, Al Bughaili M, Lin A, Sahai I, Bahena P, Reichert S, Loh A, Wright G, Liu J, Rahikkala E, Pivnick E, Choudhri A, Krüger U, Zemojtel T, van Ravenswaaij-Arts C, Mostafavi R, Stolte-Dijkstra I, Symoens S, Pajunen L, Al-Gazali L, Meierhofer D, Robinson P, Mundlos S, Villarroel C, Byers P, Masri A, Robertson S, Schwarze U, Callewaert B, Reversade B, Kornak U. Recurrent De Novo Mutations Affecting Residue Arg138 of Pyrroline-5-Carboxylate Synthase Cause a Progeroid Form of Autosomal-Dominant Cutis Laxa. Am J Hum Genet 2015; 97:483-92. [PMID: 26320891 DOI: 10.1016/j.ajhg.2015.08.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/03/2015] [Indexed: 11/24/2022] Open
Abstract
Progeroid disorders overlapping with De Barsy syndrome (DBS) are collectively denoted as autosomal-recessive cutis laxa type 3 (ARCL3). They are caused by biallelic mutations in PYCR1 or ALDH18A1, encoding pyrroline-5-carboxylate reductase 1 and pyrroline-5-carboxylate synthase (P5CS), respectively, which both operate in the mitochondrial proline cycle. We report here on eight unrelated individuals born to non-consanguineous families clinically diagnosed with DBS or wrinkly skin syndrome. We found three heterozygous mutations in ALDH18A1 leading to amino acid substitutions of the same highly conserved residue, Arg138 in P5CS. A de novo origin was confirmed in all six probands for whom parental DNA was available. Using fibroblasts from affected individuals and heterologous overexpression, we found that the P5CS-p.Arg138Trp protein was stable and able to interact with wild-type P5CS but showed an altered sub-mitochondrial distribution. A reduced size upon native gel electrophoresis indicated an alteration of the structure or composition of P5CS mutant complex. Furthermore, we found that the mutant cells had a reduced P5CS enzymatic activity leading to a delayed proline accumulation. In summary, recurrent de novo mutations, affecting the highly conserved residue Arg138 of P5CS, cause an autosomal-dominant form of cutis laxa with progeroid features. Our data provide insights into the etiology of cutis laxa diseases and will have immediate impact on diagnostics and genetic counseling.
Collapse
|
64
|
Vanakker O, Callewaert B, Malfait F, Coucke P. The Genetics of Soft Connective Tissue Disorders. Annu Rev Genomics Hum Genet 2015; 16:229-55. [DOI: 10.1146/annurev-genom-090314-050039] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Olivier Vanakker
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Bert Callewaert
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Fransiska Malfait
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Paul Coucke
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium;
| |
Collapse
|
65
|
Elevation of HO-1 Expression Mitigates Intestinal Ischemia-Reperfusion Injury and Restores Tight Junction Function in a Rat Liver Transplantation Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:986075. [PMID: 26064429 PMCID: PMC4441991 DOI: 10.1155/2015/986075] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 04/26/2015] [Indexed: 12/17/2022]
Abstract
Aims. This study was aimed at investigating whether elevation of heme oxygenase-1 (HO-1) expression could lead to restoring intestinal tight junction (TJ) function in a rat liver transplantation model. Methods. Intestinal mucosa injury was induced by orthotopic autologous liver transplantation (OALT) on male Sprague-Dawley rats. Hemin (a potent HO-1 activator) and zinc-protoporphyrin (ZnPP, a HO-1 competitive inhibitor), were separately administered in selected groups before OALT. The serum and intestinal mucosa samples were collected at 8 hours after the operation for analysis. Results. Hemin pretreatment significantly reduced the inflammation and oxidative stress in the mucosal tissue after OALT by elevating HO-1 protein expression, while ZnPP pretreatment aggravated the OALT mucosa injury. Meanwhile, the restriction on the expression of tight junction proteins zonula occludens-1 and occludin was removed after hemin pretreatment. These molecular events led to significant improvement on intestinal barrier function, which was proved to be through increasing nuclear translocation of nuclear factor-E2-related factor 2 (Nrf2) and reducing nuclear translocation of nuclear factor kappa-B (NF-κB) in intestinal injured mucosa. Summary. Our study demonstrated that elevation of HO-1 expression reduced the OALT-induced intestinal mucosa injury and TJ dysfunction. The HO-1 protective function was likely mediated through its effects of anti-inflammation and antioxidative stress.
Collapse
|
66
|
Abstract
The LTBPs (or latent transforming growth factor β binding proteins) are important components of the extracellular matrix (ECM) that interact with fibrillin microfibrils and have a number of different roles in microfibril biology. There are four LTBPs isoforms in the human genome (LTBP-1, -2, -3, and -4), all of which appear to associate with fibrillin and the biology of each isoform is reviewed here. The LTBPs were first identified as forming latent complexes with TGFβ by covalently binding the TGFβ propeptide (LAP) via disulfide bonds in the endoplasmic reticulum. LAP in turn is cleaved from the mature TGFβ precursor in the trans-golgi network but LAP and TGFβ remain strongly bound through non-covalent interactions. LAP, TGFβ, and LTBP together form the large latent complex (LLC). LTBPs were originally thought to primarily play a role in maintaining TGFβ latency and targeting the latent growth factor to the extracellular matrix (ECM), but it has also been shown that LTBP-1 participates in TGFβ activation by integrins and may also regulate activation by proteases and other factors. LTBP-3 appears to have a role in skeletal formation including tooth development. As well as having important functions in TGFβ regulation, TGFβ-independent activities have recently been identified for LTBP-2 and LTBP-4 in stabilizing microfibril bundles and regulating elastic fiber assembly.
Collapse
|
67
|
Su CT, Huang JW, Chiang CK, Lawrence EC, Levine KL, Dabovic B, Jung C, Davis EC, Madan-Khetarpal S, Urban Z. Latent transforming growth factor binding protein 4 regulates transforming growth factor beta receptor stability. Hum Mol Genet 2015; 24:4024-36. [PMID: 25882708 DOI: 10.1093/hmg/ddv139] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 04/13/2015] [Indexed: 01/17/2023] Open
Abstract
Mutations in the gene for the latent transforming growth factor beta binding protein 4 (LTBP4) cause autosomal recessive cutis laxa type 1C. To understand the molecular disease mechanisms of this disease, we investigated the impact of LTBP4 loss on transforming growth factor beta (TGFβ) signaling. Despite elevated extracellular TGFβ activity, downstream signaling molecules of the TGFβ pathway, including pSMAD2 and pERK, were down-regulated in LTBP4 mutant human dermal fibroblasts. In addition, TGFβ receptors 1 and 2 (TGFBR1 and TGFBR2) were reduced at the protein but not at the ribonucleic acid level. Treatment with exogenous TGFβ1 led to an initially rapid increase in SMAD2 phosphorylation followed by a sustained depression of phosphorylation and receptor abundance. In mutant cells TGFBR1 was co-localized with lysosomes. Treatment with a TGFBR1 kinase inhibitor, endocytosis inhibitors or a lysosome inhibitor, normalized the levels of TGFBR1 and TGFBR2. Co-immunoprecipitation demonstrated a molecular interaction between LTBP4 and TGFBR2. Knockdown of LTBP4 reduced TGFβ receptor abundance and signaling in normal cells and supplementation of recombinant LTBP4 enhanced these measures in mutant cells. In a mouse model of Ltbp4 deficiency, reduced TGFβ signaling and receptor levels were normalized upon TGFBR1 kinase inhibitor treatment. Our results show that LTBP4 interacts with TGFBR2 and stabilizes TGFβ receptors by preventing their endocytosis and lysosomal degradation in a ligand-dependent and receptor kinase activity-dependent manner. These findings identify LTBP4 as a key molecule required for the stability of the TGFβ receptor complex, and a new mechanism by which the extracellular matrix regulates cytokine receptor signaling.
Collapse
Affiliation(s)
- Chi-Ting Su
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Crabtree Hall A300, Pittsburgh, PA 15261, USA
| | - Jenq-Wen Huang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Medical College and Hospital, Taipei 100, Taiwan
| | - Chih-Kang Chiang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Medical College and Hospital, Taipei 100, Taiwan
| | - Elizabeth C Lawrence
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Crabtree Hall A300, Pittsburgh, PA 15261, USA
| | - Kara L Levine
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Crabtree Hall A300, Pittsburgh, PA 15261, USA
| | - Branka Dabovic
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | - Christine Jung
- Practice of Human Genetics, Karlsruhe 76133, Germany, Synlab MVZ Human Genetics, Mannheim 68163, Germany
| | - Elaine C Davis
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 2B2, Canada and
| | - Suneeta Madan-Khetarpal
- Division of Medical Genetics, Children's Hospital of Pittsburgh of UMPC, Pittsburgh, PA 15224, USA
| | - Zsolt Urban
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto Street, Crabtree Hall A300, Pittsburgh, PA 15261, USA,
| |
Collapse
|
68
|
Bultmann-Mellin I, Conradi A, Maul AC, Dinger K, Wempe F, Wohl AP, Imhof T, Wunderlich FT, Bunck AC, Nakamura T, Koli K, Bloch W, Ghanem A, Heinz A, von Melchner H, Sengle G, Sterner-Kock A. Modeling autosomal recessive cutis laxa type 1C in mice reveals distinct functions for Ltbp-4 isoforms. Dis Model Mech 2015; 8:403-15. [PMID: 25713297 PMCID: PMC4381339 DOI: 10.1242/dmm.018960] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/16/2015] [Indexed: 01/03/2023] Open
Abstract
Recent studies have revealed an important role for LTBP-4 in elastogenesis. Its mutational inactivation in humans causes autosomal recessive cutis laxa type 1C (ARCL1C), which is a severe disorder caused by defects of the elastic fiber network. Although the human gene involved in ARCL1C has been discovered based on similar elastic fiber abnormalities exhibited by mice lacking the short Ltbp-4 isoform (Ltbp4S(-/-)), the murine phenotype does not replicate ARCL1C. We therefore inactivated both Ltbp-4 isoforms in the mouse germline to model ARCL1C. Comparative analysis of Ltbp4S(-/-) and Ltbp4-null (Ltbp4(-/-)) mice identified Ltbp-4L as an important factor for elastogenesis and postnatal survival, and showed that it has distinct tissue expression patterns and specific molecular functions. We identified fibulin-4 as a previously unknown interaction partner of both Ltbp-4 isoforms and demonstrated that at least Ltbp-4L expression is essential for incorporation of fibulin-4 into the extracellular matrix (ECM). Overall, our results contribute to the current understanding of elastogenesis and provide an animal model of ARCL1C.
Collapse
Affiliation(s)
- Insa Bultmann-Mellin
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Anne Conradi
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Alexandra C Maul
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Katharina Dinger
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany. Department of Pediatrics and Adolescent Medicine, Medical Faculty, University of Cologne, 50937 Cologne, Germany
| | - Frank Wempe
- Department of Molecular Hematology, University of Frankfurt Medical School, 60590 Frankfurt am Main, Germany
| | - Alexander P Wohl
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Thomas Imhof
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany. Institute for Dental Research and Oral Musculoskeletal Biology, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - F Thomas Wunderlich
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany. Max Planck Institute for Metabolism Research, 50931 Cologne, Germany. Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Alexander C Bunck
- Department of Radiology, Medical Faculty, University of Cologne, 50937 Cologne, Germany
| | - Tomoyuki Nakamura
- Department of Pharmacology, Kansai Medical University, Osaka 570-8506, Japan
| | - Katri Koli
- Research Programs Unit and Transplantation Laboratory, Haartman Institute, University of Helsinki, 00014 Helsinki, Finland
| | - Wilhelm Bloch
- Institute of Cardiology and Sports Medicine, German Sport University Cologne, 50933 Cologne, Germany
| | - Alexander Ghanem
- Department of Medicine/Cardiology, University of Bonn, 53127 Bonn, Germany
| | - Andrea Heinz
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Harald von Melchner
- Department of Molecular Hematology, University of Frankfurt Medical School, 60590 Frankfurt am Main, Germany
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, 50931 Cologne, Germany. Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Anja Sterner-Kock
- Center for Experimental Medicine, Medical Faculty, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
69
|
Dabovic B, Robertson IB, Zilberberg L, Vassallo M, Davis EC, Rifkin DB. Function of latent TGFβ binding protein 4 and fibulin 5 in elastogenesis and lung development. J Cell Physiol 2015; 230:226-36. [PMID: 24962333 DOI: 10.1002/jcp.24704] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/20/2014] [Indexed: 01/20/2023]
Abstract
Mice deficient in Latent TGFβ Binding Protein 4 (Ltbp4) display a defect in lung septation and elastogenesis. The lung septation defect is normalized by genetically decreasing TGFβ2 levels. However, the elastic fiber assembly is not improved in Tgfb2(-/-) ;Ltbp4S(-/-) compared to Ltbp4S(-/-) lungs. We found that decreased levels of TGFβ1 or TGFβ3 did not improve lung septation indicating that the TGFβ isoform elevated in Ltbp4S(-/-) lungs is TGFβ2. Expression of a form of Ltbp4 that could not bind latent TGFβ did not affect lung phenotype indicating that normal lung development does not require the formation of LTBP4-latent TGFβ complexes. Therefore, the change in TGFβ-level in the lungs is not directly related to Ltbp4 deficiency but probably is a consequence of changes in the extracellular matrix. Interestingly, combination of the Ltbp4S(-/-) mutation with a fibulin-5 null mutant in Fbln5(-/-) ;Ltbp4S(-/-) mice improves the lung septation compared to Ltbp4S(-/-) lungs. Large globular elastin aggregates characteristic for Ltbp4S(-/-) lungs do not form in Fbln5(-/-) ;Ltbp4S(-/-) lungs and EM studies showed that elastic fibers in Fbln5(-/-) ;Ltbp4S(-/-) lungs resemble those found in Fbln5(-/-) mice. These results are consistent with a role for TGFβ2 in lung septation and for Ltbp4 in regulating fibulin-5 dependent elastic fiber assembly.
Collapse
Affiliation(s)
- Branka Dabovic
- Departments of Cell Biology, New York University Medical Center, 550 First Avenue, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
70
|
Abstract
Congenital diaphragmatic hernia (CDH) is a moderately prevalent birth defect that, despite advances in neonatal care, is still a significant cause of infant death, and surviving patients have significant morbidity. The goal of ongoing research to elucidate the genetic causes of CDH is to develop better treatment and ultimately prevention. CDH is a complex developmental defect that is etiologically heterogeneous. This review summarizes the recurrent genetic causes of CDH including aneuploidies, chromosome copy number variants, and single gene mutations. It also discusses strategies for genetic evaluation and genetic counseling in an era of rapidly evolving technologies in clinical genetic diagnostics.
Collapse
Affiliation(s)
| | | | - Wendy K. Chung
- Corresponding author. Address: Division of Molecular Genetics, Department of Pediatrics, Columbia University Medical Center, 1150 St Nicholas Avenue, Room 620, New York, NY 10032, USA. Tel.: +1 212-851-5313; fax: +1 212-851-5306. (W.K. Chung)
| |
Collapse
|
71
|
Gardeitchik T, Mohamed M, Fischer B, Lammens M, Lefeber D, Lace B, Parker M, Kim KJ, Lim BC, Häberle J, Garavelli L, Jagadeesh S, Kariminejad A, Guerra D, Leão M, Keski-Filppula R, Brunner H, Nijtmans L, van den Heuvel B, Wevers R, Kornak U, Morava E. Clinical and biochemical features guiding the diagnostics in neurometabolic cutis laxa. Eur J Hum Genet 2014; 22:888-95. [PMID: 23963297 PMCID: PMC4060105 DOI: 10.1038/ejhg.2013.154] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 06/11/2013] [Accepted: 06/12/2013] [Indexed: 01/01/2023] Open
Abstract
Patients with cutis laxa (CL) have wrinkled, sagging skin with decreased elasticity. Skin symptoms are associated with variable systemic involvement. The most common, genetically highly heterogeneous form of autosomal recessive CL, ARCL2, is frequently associated with variable metabolic and neurological symptoms. Progeroid symptoms, dysmorphic features, hypotonia and psychomotor retardation are highly overlapping in the early phase of these disorders. This makes the genetic diagnosis often challenging. In search for discriminatory symptoms, we prospectively evaluated clinical, neurologic, metabolic and genetic features in our patient cohort referred for suspected ARCL. From a cohort of 26 children, we confirmed mutations in genes associated with ARCL in 16 children (14 probands), including 12 novel mutations. Abnormal glycosylation and gyration abnormalities were mostly, but not always associated with ATP6V0A2 mutations. Epilepsy was most common in ATP6V0A2 defects. Corpus callosum dysgenesis was associated with PYCR1 and ALDH18A1 mutations. Dystonic posturing was discriminatory for PYCR1 and ALDH18A1 defects. Metabolic markers of mitochondrial dysfunction were found in one patient with PYCR1 mutations. So far unreported white matter abnormalities were found associated with GORAB and RIN2 mutations. We describe a large cohort of CL patients with neurologic involvement. Migration defects and corpus callosum hypoplasia were not always diagnostic for a specific genetic defect in CL. All patients with ATP6V0A2 defects had abnormal glycosylation. To conclude, central nervous system and metabolic abnormalities were discriminatory in this genetically heterogeneous group, although not always diagnostic for a certain genetic defect in CL.
Collapse
Affiliation(s)
- Thatjana Gardeitchik
- Department of Pediatrics, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Miski Mohamed
- Department of Pediatrics, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Björn Fischer
- Institute of Medical Genetics and Human Genetics, Charité Universitätsmedizin, Berlin, Germany
| | - Martin Lammens
- Department of Pathology, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Dirk Lefeber
- Department of Neurology, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Baiba Lace
- Medical Genetics Clinic, Children's Clinical University Hospital, Riga, Latvia
| | - Michael Parker
- Sheffield Clinical Genetics Service, Sheffield Children's Hospital, Sheffield, UK
| | - Ki-Joong Kim
- Department of Pediatrics, Seoul National University Hospital, Seoul, South Korea
| | - Bing C Lim
- Department of Pediatrics, Seoul National University Hospital, Seoul, South Korea
| | - Johannes Häberle
- Department of Pediatrics, University Children's Hospital, Zürich, Switzerland
| | - Livia Garavelli
- Clinical Genetics Unit, Obstetric and Pediatric Department, Santa Maria Nuova Hospital IRCCS, Reggio Emilia, Italy
| | | | | | - Deanna Guerra
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Michel Leão
- Pediatric Neurology Unit and Neurogenetics Unit, Hospital S João, Porto, Portugal
| | | | - Han Brunner
- Department of Human Genetics, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Leo Nijtmans
- Department of Pediatrics, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Bert van den Heuvel
- Department of Pediatrics, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
- Laboratory for Genetic Endocrine and Metabolic Diseases, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Ron Wevers
- Department of Pediatrics, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
- Laboratory for Genetic Endocrine and Metabolic Diseases, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Uwe Kornak
- Institute of Medical Genetics and Human Genetics, Charité Universitätsmedizin, Berlin, Germany
- FG Development and Disease, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Eva Morava
- Department of Pediatrics, Institute for Metabolic and Genetic Disease, Radboud University Medical Centre, Nijmegen, The Netherlands
- Hayward Genetics Center, Tulane University Medical Center, New Orleans, LA, USA
| |
Collapse
|
72
|
Kantaputra PN, Kaewgahya M, Wiwatwongwana A, Wiwatwongwana D, Sittiwangkul R, Iamaroon A, Dejkhamron P. Cutis laxa with pulmonary emphysema, conjunctivochalasis, nasolacrimal duct obstruction, abnormal hair, and a novel FBLN5 mutation. Am J Med Genet A 2014; 164A:2370-7. [PMID: 24962763 DOI: 10.1002/ajmg.a.36630] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Accepted: 05/16/2014] [Indexed: 12/15/2022]
Abstract
We report on a 4-year-old girl with autosomal recessive cutis laxa, type IA, or pulmonary emphysema type (ARCL1A; OMIM #219100), with loose and wrinkled skin, mitral and tricuspid valve prolapse, conjunctivochalasis, obstructed nasolacrimal ducts, hypoplastic maxilla, and early childhood-onset pulmonary emphysema. Mutation analysis of FBLN5 showed a homozygous c.432C>G missense mutation, and heterozygosity in the parents. This is predicted to cause amino acid substitution p.Cys144Trp. Conjunctivochalasis or redundant folds of conjunctiva and obstructed nasolacrimal ducts have not been reported to be associated with FBLN5 mutations. Histopathological study of the conjunctival biopsy showed that most blood vessels had normal elastic fibers. The gingiva appeared normal, but histologically elastic fibers were defective. Scanning electron micrography of scalp hair demonstrated hypoplastic hair follicles. The cuticles appear intact underneath the filamentous meshwork.
Collapse
Affiliation(s)
- Piranit Nik Kantaputra
- Center of Excellence in Medical Genetics Research, Chiang Mai University, Chiang Mai, Thailand; Division of Pediatric Dentistry, Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand; Craniofacial Genetics Laboratory, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand; Dentaland Clinic, Chiang Mai, Thailand
| | | | | | | | | | | | | |
Collapse
|
73
|
The function of elastic fibers in the arteries: beyond elasticity. ACTA ACUST UNITED AC 2014; 62:79-83. [PMID: 24679588 DOI: 10.1016/j.patbio.2014.02.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 02/24/2014] [Indexed: 11/23/2022]
Abstract
The main components of elastic fibers, elastin and fibrillin-containing microfibrils play a structural and mechanical role in the arteries and their essential function is to provide elasticity and resilience to the tissues. However, through control of the quiescent contractile phenotype of arterial smooth muscle cells, elastin also acts as an autocrine factor and, via the binding of 'latent transforming growth factor (TGF)-β binding protein (LTBP) - latency-associated peptide (LAP) - TGF-β' complexes, fibrillins regulate the activation and availability of TGF-βs. These recent discoveries are detailed in this review.
Collapse
|
74
|
Handley MT, Mégarbané A, Meynert AM, Brown S, Freyer E, Taylor MS, Jackson IJ, Aligianis IA. Loss of ALDH18A1 function is associated with a cellular lipid droplet phenotype suggesting a link between autosomal recessive cutis laxa type 3A and Warburg Micro syndrome. Mol Genet Genomic Med 2014; 2:319-25. [PMID: 25077174 PMCID: PMC4113272 DOI: 10.1002/mgg3.70] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/17/2014] [Accepted: 01/29/2014] [Indexed: 12/16/2022] Open
Abstract
Autosomal recessive cutis laxa type 3A is caused by mutations in ALDH18A1, a gene encoding the mitochondrial enzyme Δ1-pyrroline-5-carboxylate synthase (P5CS). It is a rare disorder with only six pathogenic mutations and 10 affected individuals from five families previously described in the literature. Here we report the identification of novel compound heterozygous missense mutations in two affected siblings from a Lebanese family by whole-exome sequencing. The mutations alter a conserved C-terminal domain of the encoded protein and reduce protein stability as determined through Western blot analysis of patient fibroblasts. Patient fibroblasts exhibit a lipid droplet phenotype similar to that recently reported in Warburg Micro syndrome, a disorder with similar features but hitherto unrelated cellular etiology.
Collapse
Affiliation(s)
- Mark T Handley
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh Edinburgh, UK
| | - André Mégarbané
- Institut Médical Jérôme Lejeune et Fondation Jérome Lejeune Paris, France ; Unité de Génétique Médicale, Faculté de Médecine, Université Saint-Joseph Beirut, Lebanon
| | - Alison M Meynert
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh Edinburgh, UK
| | - Stephen Brown
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh Edinburgh, UK
| | - Elisabeth Freyer
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh Edinburgh, UK
| | - Martin S Taylor
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh Edinburgh, UK
| | - Ian J Jackson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh Edinburgh, UK
| | - Irene A Aligianis
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh Edinburgh, UK
| |
Collapse
|
75
|
Ryan DM, Vincent TL, Salit J, Walters MS, Agosto-Perez F, Shaykhiev R, Strulovici-Barel Y, Downey RJ, Buro-Auriemma LJ, Staudt MR, Hackett NR, Mezey JG, Crystal RG. Smoking dysregulates the human airway basal cell transcriptome at COPD risk locus 19q13.2. PLoS One 2014; 9:e88051. [PMID: 24498427 PMCID: PMC3912203 DOI: 10.1371/journal.pone.0088051] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/03/2014] [Indexed: 11/19/2022] Open
Abstract
Genome-wide association studies (GWAS) and candidate gene studies have identified a number of risk loci associated with the smoking-related disease COPD, a disorder that originates in the airway epithelium. Since airway basal cell (BC) stem/progenitor cells exhibit the earliest abnormalities associated with smoking (hyperplasia, squamous metaplasia), we hypothesized that smoker BC have a dysregulated transcriptome, enriched, in part, at known GWAS/candidate gene loci. Massive parallel RNA sequencing was used to compare the transcriptome of BC purified from the airway epithelium of healthy nonsmokers (n = 10) and healthy smokers (n = 7). The chromosomal location of the differentially expressed genes was compared to loci identified by GWAS to confer risk for COPD. Smoker BC have 676 genes differentially expressed compared to nonsmoker BC, dominated by smoking up-regulation. Strikingly, 166 (25%) of these genes are located on chromosome 19, with 13 localized to 19q13.2 (p<10−4 compared to chance), including 4 genes (NFKBIB, LTBP4, EGLN2 and TGFB1) associated with risk for COPD. These observations provide the first direct connection between known genetic risks for smoking-related lung disease and airway BC, the population of lung cells that undergo the earliest changes associated with smoking.
Collapse
Affiliation(s)
- Dorothy M. Ryan
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Thomas L. Vincent
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Jacqueline Salit
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Matthew S. Walters
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Francisco Agosto-Perez
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Renat Shaykhiev
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Yael Strulovici-Barel
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Robert J. Downey
- Thoracic Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Lauren J. Buro-Auriemma
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Michelle R. Staudt
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Neil R. Hackett
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
| | - Jason G. Mezey
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Ronald G. Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
76
|
Greally MT, Kalis NN, Agab W, Ardati K, Giurgea S, Kornak U, Van Maldergem L. Autosomal recessive cutis laxa type 2A (ARCL2A) mimicking Ehlers-Danlos syndrome by its dermatological manifestations: Report of three affected patients. Am J Med Genet A 2014; 164A:1245-53. [DOI: 10.1002/ajmg.a.36411] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 11/10/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Marie T. Greally
- National Centre for Medical Genetics; Our Lady's Children's Hospital, Crumlin; Dublin Ireland
- Bahrain Defence Forces Hospital; Department of Paediatric Cardiology; Riffa Kingdom of Bahrain
| | - Neale N. Kalis
- Bahrain Defence Forces Hospital; Department of Paediatric Cardiology; Riffa Kingdom of Bahrain
| | - Wahid Agab
- Bahrain Defence Forces Hospital; Department of Paediatric Cardiology; Riffa Kingdom of Bahrain
| | - Kasim Ardati
- Bahrain Specialist Hospital; Juffair Kingdom of Bahrain
| | - Sanda Giurgea
- Department of Neurology; CHU Tivoli; La Louvière Belgium
| | - Uwe Kornak
- Institute of Medical and Human Genetics; Charité-Universitaetsmedizin Berlin; Berlin Germany
| | | |
Collapse
|
77
|
Van Asbeck E, Wolthuis DF, Mohamed M, Wevers RA, Korenke CG, Gardeitchik T, Morava E. A novel phenotype associated with cutis laxa, abnormal fat distribution, cardiomyopathy and cataract. Am J Med Genet A 2014; 164A:1049-55. [DOI: 10.1002/ajmg.a.36392] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 11/07/2013] [Indexed: 11/06/2022]
Affiliation(s)
- Ellyze Van Asbeck
- Department of Pediatrics; Radboud University Nijmegen Medical Center; Nijmegen Gelderland Netherlands
- Hayward Genetics Center and Department of Pediatrics; Tulane University Medical School; New Orleans Louisiana
| | - David F.G.J. Wolthuis
- Department of Pediatrics; Radboud University Nijmegen Medical Center; Nijmegen Gelderland Netherlands
- Hayward Genetics Center and Department of Pediatrics; Tulane University Medical School; New Orleans Louisiana
| | - Miski Mohamed
- Department of Pediatrics; Radboud University Nijmegen Medical Center; Nijmegen Gelderland Netherlands
| | - Ron A. Wevers
- Laboratory of Genetic, Endocrine and Metabolic Disease; Radboud University Nijmegen Medical Center; Nijmegen Gelderland Netherlands
| | | | - Thatjana Gardeitchik
- Department of Pediatrics; Radboud University Nijmegen Medical Center; Nijmegen Gelderland Netherlands
| | - Eva Morava
- Department of Pediatrics; Radboud University Nijmegen Medical Center; Nijmegen Gelderland Netherlands
- Hayward Genetics Center and Department of Pediatrics; Tulane University Medical School; New Orleans Louisiana
| |
Collapse
|
78
|
|
79
|
Chandramouli A, Simundza J, Pinderhughes A, Hiremath M, Droguett G, Frendewey D, Cowin P. Ltbp1L is focally induced in embryonic mammary mesenchyme, demarcates the ductal luminal lineage and is upregulated during involution. Breast Cancer Res 2013; 15:R111. [PMID: 24262428 PMCID: PMC3978911 DOI: 10.1186/bcr3578] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 10/31/2013] [Indexed: 11/10/2022] Open
Abstract
Introduction Latent TGFβ binding proteins (LTBPs) govern TGFβ presentation and activation and are important for elastogenesis. Although TGFβ is well-known as a tumor suppressor and metastasis promoter, and LTBP1 is elevated in two distinct breast cancer metastasis signatures, LTBPs have not been studied in the normal mammary gland. Methods To address this we have examined Ltbp1 promoter activity throughout mammary development using an Ltbp1L-LacZ reporter as well as expression of both Ltbp1L and 1S mRNA and protein by qRT-PCR, immunofluorescence and flow cytometry. Results Our data show that Ltbp1L is transcribed coincident with lumen formation, providing a rare marker distinguishing ductal from alveolar luminal lineages. Ltbp1L and Ltbp1S are silent during lactation but robustly induced during involution, peaking at the stage when the remodeling process becomes irreversible. Ltbp1L is also induced within the embryonic mammary mesenchyme and maintained within nipple smooth muscle cells and myofibroblasts. Ltbp1 protein exclusively ensheaths ducts and side branches. Conclusions These data show Ltbp1 is transcriptionally regulated in a dynamic manner that is likely to impose significant spatial restriction on TGFβ bioavailability during mammary development. We hypothesize that Ltbp1 functions in a mechanosensory capacity to establish and maintain ductal luminal cell fate, support and detect ductal distension, trigger irreversible involution, and facilitate nipple sphincter function.
Collapse
|
80
|
Dimopoulou A, Fischer B, Gardeitchik T, Schröter P, Kayserili H, Schlack C, Li Y, Brum JM, Barisic I, Castori M, Spaich C, Fletcher E, Mahayri Z, Bhat M, Girisha KM, Lachlan K, Johnson D, Phadke S, Gupta N, Simandlova M, Kabra M, David A, Nijtmans L, Chitayat D, Tuysuz B, Brancati F, Mundlos S, Van Maldergem L, Morava E, Wollnik B, Kornak U. Genotype-phenotype spectrum of PYCR1-related autosomal recessive cutis laxa. Mol Genet Metab 2013; 110:352-61. [PMID: 24035636 DOI: 10.1016/j.ymgme.2013.08.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 08/12/2013] [Accepted: 08/14/2013] [Indexed: 10/26/2022]
Abstract
Autosomal recessive cutis laxa type 2B (ARCL2B; OMIM # 612940) is a segmental progeroid disorder caused by mutations in PYCR1 encoding pyrroline-5-carboxylate reductase 1, which is part of the conserved proline de novo synthesis pathway. Here we describe 33 patients with PYCR1-related ARCL from 27 families with initial diagnoses varying between wrinkly skin syndrome, gerodermia osteodysplastica, De Barsy syndrome or more severe progeria syndromes. Given the difficult differential diagnosis of ARCL syndromes we performed a systematic comparison of clinical features of PYCR1-related ARCL. Intrauterine growth retardation, a characteristic triangular facial gestalt, psychomotor retardation, and hypotonia were the most relevant distinctive hallmarks of ARCL due to proline de novo synthesis defects. Corneal clouding or cataracts, athetoid movements, and finger contractures were rather rare features, but had a high predictive value. In our cohort we identified 20 different PYCR1 mutations of which seven were novel. Most of the mutations accumulated in exons 4 to 6. Missense alterations of highly conserved residues were most frequent followed by splice site changes and a single nonsense mutation. Analysis of genotype-phenotype correlation revealed that patients with mutations in the first two exons had lower average clinical scores and absent or only mild intellectual disability. Structural analyses predicted interference with PYCR1 multimerization for a subset of missense mutations. These findings have implications for the clinics as well as the pathomechanism of PYCR1-related ARCL.
Collapse
Affiliation(s)
- Aikaterini Dimopoulou
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Augustenburger Platz 1, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Ceco E, McNally EM. Modifying muscular dystrophy through transforming growth factor-β. FEBS J 2013; 280:4198-209. [PMID: 23551962 PMCID: PMC3731412 DOI: 10.1111/febs.12266] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 03/11/2013] [Accepted: 03/20/2013] [Indexed: 12/31/2022]
Abstract
Muscular dystrophy arises from ongoing muscle degeneration and insufficient regeneration. This imbalance leads to loss of muscle, with replacement by scar or fibrotic tissue, resulting in muscle weakness and, eventually, loss of muscle function. Human muscular dystrophy is characterized by a wide range of disease severity, even when the same genetic mutation is present. This variability implies that other factors, both genetic and environmental, modify the disease outcome. There has been an ongoing effort to define the genetic and molecular bases that influence muscular dystrophy onset and progression. Modifier genes for muscle disease have been identified through both candidate gene approaches and genome-wide surveys. Multiple lines of experimental evidence have now converged on the transforming growth factor-β (TGF-β) pathway as a modifier for muscular dystrophy. TGF-β signaling is upregulated in dystrophic muscle as a result of a destabilized plasma membrane and/or an altered extracellular matrix. Given the important biological role of the TGF-β pathway, and its role beyond muscle homeostasis, we review modifier genes that alter the TGF-β pathway and approaches to modulate TGF-β activity to ameliorate muscle disease.
Collapse
Affiliation(s)
- Ermelinda Ceco
- Committee on Cell Physiology, University of Chicago, IL 60637, USA
| | | |
Collapse
|
82
|
Abstract
Elastic fibres are insoluble components of the extracellular matrix of dynamic connective tissues such as skin, arteries, lungs and ligaments. They are laid down during development, and comprise a cross-linked elastin core within a template of fibrillin-based microfibrils. Their function is to endow tissues with the property of elastic recoil, and they also regulate the bioavailability of transforming growth factor β. Severe heritable elastic fibre diseases are caused by mutations in elastic fibre components; for example, mutations in elastin cause supravalvular aortic stenosis and autosomal dominant cutis laxa, mutations in fibrillin-1 cause Marfan syndrome and Weill–Marchesani syndrome, and mutations in fibulins-4 and -5 cause autosomal recessive cutis laxa. Acquired elastic fibre defects include dermal elastosis, whereas inflammatory damage to fibres contributes to pathologies such as pulmonary emphysema and vascular disease. This review outlines the latest understanding of the composition and assembly of elastic fibres, and describes elastic fibre diseases and current therapeutic approaches.
Collapse
|
83
|
Urban Z, Davis EC. Cutis laxa: intersection of elastic fiber biogenesis, TGFβ signaling, the secretory pathway and metabolism. Matrix Biol 2013; 33:16-22. [PMID: 23954411 DOI: 10.1016/j.matbio.2013.07.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 07/08/2013] [Accepted: 07/09/2013] [Indexed: 12/11/2022]
Abstract
Cutis laxa (CL), a disease characterized by redundant and inelastic skin, displays extensive locus heterogeneity. Together with geroderma osteodysplasticum and arterial tortuosity syndrome, which show phenotypic overlap with CL, eleven CL-related genes have been identified to date, which encode proteins within 3 groups. Elastin, fibulin-4, fibulin-5 and latent transforming growth factor-β-binding protein 4 are secreted proteins which form elastic fibers and are involved in the sequestration and subsequent activation of transforming growth factor-β (TGFβ). Proteins within the second group, localized to the secretory pathway, perform transport and membrane trafficking functions necessary for the modification and secretion of elastic fiber components. Key proteins include a subunit of the vacuolar-type proton pump, which ensures the efficient secretion of tropoelastin, the precursor or elastin. A copper transporter is required for the activity of lysyl oxidases, which crosslink collagen and elastin. A Rab6-interacting goglin recruits kinesin motors to Golgi-vesicles facilitating the transport from the Golgi to the plasma membrane. The Rab and Ras interactor 2 regulates the activity of Rab5, a small guanosine triphosphatase essential for the endocytosis of various cell surface receptors, including integrins. Proteins of the third group related to CL perform metabolic functions within the mitochondria, inhibiting the accumulation of reactive oxygen species. Two of these proteins catalyze subsequent steps in the conversion of glutamate to proline. The third transports dehydroascorbate into mitochondria. Recent studies on CL-related proteins highlight the intricate connections among membrane trafficking, metabolism, extracellular matrix assembly, and TGFβ signaling.
Collapse
Affiliation(s)
- Zsolt Urban
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| | - Elaine C Davis
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, H3A 0C7 Canada
| |
Collapse
|
84
|
Merla G, Brunetti-Pierri N, Piccolo P, Micale L, Loviglio MN. Supravalvular aortic stenosis: elastin arteriopathy. ACTA ACUST UNITED AC 2013; 5:692-6. [PMID: 23250899 DOI: 10.1161/circgenetics.112.962860] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supravalvular aortic stenosis is a systemic elastin (ELN) arteriopathy that disproportionately affects the supravalvular aorta. ELN arteriopathy may be present in a nonsyndromic condition or in syndromic conditions such as Williams-Beuren syndrome. The anatomic findings include congenital narrowing of the lumen of the aorta and other arteries, such as branches of pulmonary or coronary arteries. Given the systemic nature of the disease, accurate evaluation is recommended to establish the degree and extent of vascular involvement and to plan appropriate interventions, which are indicated whenever hemodynamically significant stenoses occur. ELN arteriopathy is genetically heterogeneous and occurs as a consequence of haploinsufficiency of the ELN gene on chromosome 7q11.23, owing to either microdeletion of the entire chromosomal region or ELN point mutations. Interestingly, there is a prevalence of premature termination mutations resulting in null alleles among ELN point mutations. The identification of the genetic defect in patients with supravalvular aortic stenosis is essential for a definitive diagnosis, prognosis, and genetic counseling.
Collapse
Affiliation(s)
- Giuseppe Merla
- Medical Genetics Unit, IRCCS Casa Sollievo Della Sofferenza Hospital, San Giovanni Rotondo, Italy.
| | | | | | | | | |
Collapse
|
85
|
Van Laer L, Proost D, Loeys BL. Educational paper. Connective tissue disorders with vascular involvement: from gene to therapy. Eur J Pediatr 2013; 172:997-1005. [PMID: 22801769 PMCID: PMC3722447 DOI: 10.1007/s00431-012-1773-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 06/05/2012] [Indexed: 01/21/2023]
Abstract
Heritable connective tissue diseases comprise a heterogeneous group of multisystemic disorders that are characterized by significant morbidity and mortality. These disorders do not merely result from defects in the amount or structure of one of the components of the extracellular matrix, as the extracellular matrix also serves other functions, including sequestration of cytokines, such as transforming growth factor beta (TGFβ). Indeed, disturbed TGFβ signaling was demonstrated in several heritable connective tissue diseases, including syndromic forms such as Marfan or Loeys-Dietz syndrome and non-syndromic presentations of thoracic aortic aneurysm/dissection. Because of these findings, new therapeutic targets have been unveiled, leading to the initiation of large clinical trials with angiotensin II type 1 receptor antagonists that also have an inhibiting effect on TGFβ signaling. Here, we present an overview of the clinical characteristics, the molecular findings, and the therapeutic strategies for the currently known syndromic and non-syndromic forms of thoracic aortic aneurysm/dissection.
Collapse
Affiliation(s)
- Lut Van Laer
- Center for Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Antwerp, Edegem Belgium
| | - Dorien Proost
- Center for Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Antwerp, Edegem Belgium
| | - Bart L. Loeys
- Center for Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Antwerp, Edegem Belgium
- Department of Human Genetics, Nijmegen Centre for Molecular Life Sciences and Institute for Genetic and Metabolic Disorders, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
86
|
Bultmann I, Conradi A, Kretschmer C, Sterner-Kock A. Latent transforming growth factor β-binding protein 4 is downregulated in esophageal cancer via promoter methylation. PLoS One 2013; 8:e65614. [PMID: 23741501 PMCID: PMC3669142 DOI: 10.1371/journal.pone.0065614] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/26/2013] [Indexed: 02/07/2023] Open
Abstract
Latent transforming growth factor β-binding protein 4 (LTBP4) is an extracellular matrix molecule that is a member of important connective tissue networks and is needed for the correct folding and the secretion of TGF-β1. LTBP4 is downregulated in carcinomas of various tissues. Here we show that LTBP4 is also downregulated in adenocarcinomas and squamous cell carcinomas of the esophagus in vitro and in vivo. Re-expression of LTBP4 in esophageal cancer cell lines reduced cell migration ability, whereas cell viability and cell proliferation remained unchanged. Hypermethylation of the promoter regions of the two main human LTBP4 transcriptional forms, LTBP4L and LTBP4S, was found to be involved in LTBP4 silencing. Detailed investigations of the methylation patterns of the promoter regions of LTBP4L and LTBP4S identified GATA1, SP1, E2F4 and SMAD3 as potential transcription factors involved in LTBP4 expression. In in vitro transcription factor activity studies we discovered E2F4 as novel powerful regulator for LTBP4S expression.
Collapse
Affiliation(s)
- Insa Bultmann
- Center for Experimental Medicine, Medical Faculty, University of Cologne, Cologne, Germany
| | | | | | | |
Collapse
|
87
|
Rasouly HM, Lu W. Lower urinary tract development and disease. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2013; 5:307-42. [PMID: 23408557 PMCID: PMC3627353 DOI: 10.1002/wsbm.1212] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Congenital anomalies of the lower urinary tract (CALUT) are a family of birth defects of the ureter, the bladder, and the urethra. CALUT includes ureteral anomaliesc such as congenital abnormalities of the ureteropelvic junction (UPJ) and ureterovesical junction (UVJ), and birth defects of the bladder and the urethra such as bladder-exstrophy-epispadias complex (BEEC), prune belly syndrome (PBS), and posterior urethral valves (PUVs). CALUT is one of the most common birth defects and is often associated with antenatal hydronephrosis, vesicoureteral reflux (VUR), urinary tract obstruction, urinary tract infections (UTI), chronic kidney disease, and renal failure in children. Here, we discuss the current genetic and molecular knowledge about lower urinary tract development and genetic basis of CALUT in both human and mouse models. We provide an overview of the developmental processes leading to the formation of the ureter, the bladder, and the urethra, and different genes and signaling pathways controlling these developmental processes. Human genetic disorders that affect the ureter, the bladder and the urethra and associated gene mutations are also presented. As we are entering the postgenomic era of personalized medicine, information in this article may provide useful interpretation for the genetic and genomic test results collected from patients with lower urinary tract birth defects. With evidence-based interpretations, clinicians may provide more effective personalized therapies to patients and genetic counseling for their families.
Collapse
Affiliation(s)
- Hila Milo Rasouly
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118, USA
| | - Weining Lu
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA 02118, USA
| |
Collapse
|
88
|
Hadj-Rabia S, Callewaert BL, Bourrat E, Kempers M, Plomp AS, Layet V, Bartholdi D, Renard M, De Backer J, Malfait F, Vanakker OM, Coucke PJ, De Paepe AM, Bodemer C. Twenty patients including 7 probands with autosomal dominant cutis laxa confirm clinical and molecular homogeneity. Orphanet J Rare Dis 2013; 8:36. [PMID: 23442826 PMCID: PMC3599008 DOI: 10.1186/1750-1172-8-36] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 02/14/2013] [Indexed: 01/15/2023] Open
Abstract
Background Elastin gene mutations have been associated with a variety of phenotypes. Autosomal dominant cutis laxa (ADCL) is a rare disorder that presents with lax skin, typical facial characteristics, inguinal hernias, aortic root dilatation and pulmonary emphysema. In most patients, frameshift mutations are found in the 3’ region of the elastin gene (exons 30-34) which result in a C-terminally extended protein, though exceptions have been reported. Methods We clinically and molecularly characterized the thus far largest cohort of ADCL patients, consisting of 19 patients from six families and one sporadic patient. Results Molecular analysis showed C-terminal frameshift mutations in exon 30, 32, and 34 of the elastin gene and identified a mutational hotspot in exon 32 (c.2262delA). This cohort confirms the previously reported clinical constellation of skin laxity (100%), inguinal hernias (51%), aortic root dilatation (55%) and emphysema (37%). Conclusion ADCL is a clinically and molecularly homogeneous disorder, but intra- and interfamilial variability in the severity of organ involvement needs to be taken into account. Regular cardiovascular and pulmonary evaluations are imperative in the clinical follow-up of these patients.
Collapse
Affiliation(s)
- Smail Hadj-Rabia
- Service de Dermatologie - Centre de référence national des Maladies Génétiques à Expression Cutanée MAGEC, INSERM U781, Hôpital Necker - Enfants Malades, Université Paris V-Descartes, 149, rue de Sèvres 75743 Paris Cedex 15, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Latent TGF-β binding protein 4 promotes elastic fiber assembly by interacting with fibulin-5. Proc Natl Acad Sci U S A 2013; 110:2852-7. [PMID: 23382201 DOI: 10.1073/pnas.1215779110] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Elastic fiber assembly requires deposition of elastin monomers onto microfibrils, the mechanism of which is incompletely understood. Here we show that latent TGF-β binding protein 4 (LTBP-4) potentiates formation of elastic fibers through interacting with fibulin-5, a tropoelastin-binding protein necessary for elastogenesis. Decreased expression of LTBP-4 in human dermal fibroblast cells by siRNA treatment abolished the linear deposition of fibulin-5 and tropoelastin on microfibrils. It is notable that the addition of recombinant LTBP-4 to cell culture medium promoted elastin deposition on microfibrils without changing the expression of elastic fiber components. This elastogenic property of LTBP-4 is independent of bound TGF-β because TGF-β-free recombinant LTBP-4 was as potent an elastogenic inducer as TGF-β-bound recombinant LTBP-4. Without LTBP-4, fibulin-5 and tropoelastin deposition was discontinuous and punctate in vitro and in vivo. These data suggest a unique function for LTBP-4 during elastic fibrogenesis, making it a potential therapeutic target for elastic fiber regeneration.
Collapse
|
90
|
Shibahara K, Ota M, Horiguchi M, Yoshinaga K, Melamed J, Rifkin DB. Production of gastrointestinal tumors in mice by modulating latent TGF-β1 activation. Cancer Res 2012; 73:459-68. [PMID: 23117884 DOI: 10.1158/0008-5472.can-12-3141] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
TGF-β and its signaling pathways are important mediators in the suppression of cancers of the gastrointestinal tract. TGF-β is released from cells in a latent complex consisting of TGF-β, the TGF-β propeptide [latency associated protein (LAP)], and a latent TGF-β binding protein (LTBP). We previously generated mice in which the LTBP-binding cysteine residues in LAP TGF-β1 were mutated to serine precluding covalent interactions with LTBP. These Tgfb1(C33S/C33S) mice develop multiorgan inflammation and tumors consistent with reduced TGF-β1 activity. To test whether further reduction in active TGF-β levels would yield additional tumors and a phenotype more similar to Tgfb1(-/-) mice, we generated mice that express TGF-β1(C33S) and are deficient in either integrin β8 or TSP-1, known activators of latent TGF-β1. In addition, we generated mice that have one mutant allele and one null allele at the Tgfb1 locus, reasoning that these mice should synthesize half the total amount of TGF-β1 as Tgfb1(C33S/C33S) mice, and the amount of active TGF-β1 would be correspondingly decreased compared with Tgfb1(C33S/C33S) mice. These compound-mutant mice displayed more severe inflammation and higher tumor numbers than the parental Tgfb1(C33S/C33S) animals. The level of active TGF-β1 in compound mutant mice seemed to be decreased compared with Tgfb1(C33S/C33S) mice as determined from analyses of surrogate markers of active TGF-β, such as P-Smad2, C-Myc, KI-67, and markers of cell-cycle traverse. We conclude that these mutant mice provide a useful system for modulating TGF-β levels in a manner that determines tumor number and inflammation within the gastrointestinal tract.
Collapse
Affiliation(s)
- Kotaro Shibahara
- Department of Cell Biology, New York University Langone School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | |
Collapse
|
91
|
The Complexity of Elastic Fiber Biogenesis: The Paradigm of Cutis Laxa. J Invest Dermatol 2012; 132 Suppl 3:E12-4. [DOI: 10.1038/skinbio.2012.4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
92
|
Uitto J, Li Q, Urban Z. The complexity of elastic fibre biogenesis in the skin--a perspective to the clinical heterogeneity of cutis laxa. Exp Dermatol 2012; 22:88-92. [PMID: 23088642 DOI: 10.1111/exd.12025] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2012] [Indexed: 11/28/2022]
Abstract
Elastic fibres are critical connective tissue components providing elasticity and resilience to skin and other tissues. These fibres are composed of elastin and a number of elastin-associated microfibrillar proteins that assemble in a complex fibre network in a multi-step process. Multiple cellular processes, including mitochondrial function, specific molecules in the secretory pathways and temporally and spatially ordered production of elastic fibre components, are required for the biogenesis of functional elastic fibres. Abnormalities in these processes can lead to loss of functional elastic fibres manifesting phenotypically as a skin disease. The paradigm of elastic fibre diseases affecting the skin is cutis laxa, a clinically and genetically heterogeneous group of disorders characterized by loose and sagging skin, frequently associated with extracutaneous manifestations in the lungs and the arterial blood vessels. The complexity of cutis laxa is emphasized by the fact that as many as 10 distinct genes can harbour mutations in this and related disorders. Understanding of the pathomechanistic pathways involved in perturbed elastic fibre assembly in cutis laxa provides information potentially helpful for the development of molecular strategies towards treatment of these, currently intractable, diseases.
Collapse
Affiliation(s)
- Jouni Uitto
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | |
Collapse
|
93
|
Horiguchi M, Ota M, Rifkin DB. Matrix control of transforming growth factor-β function. J Biochem 2012; 152:321-9. [PMID: 22923731 DOI: 10.1093/jb/mvs089] [Citation(s) in RCA: 202] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The cytokine transforming growth factor-beta (TGF-β) has multiple effects in both physiological and pathological conditions. TGF-β is secreted as part of a tripartite complex from which it must be released in order to bind to its receptor. Sequestration of latent TGF-β in the extracellular matrix (ECM) is crucial for proper mobilization of the latent cytokine and its activation. However, contrary to expectation, loss-of-function mutations in genes encoding certain matrix proteins that bind TGF-β yield elevated, rather than decreased, TGF-β levels, posing a 'TGF-β paradox.' In this review, we discuss recent findings concerning the relationship of TGF-β, ECM molecules, and latent TGF-β activation and propose a model to resolve the 'TGF-β paradox.'
Collapse
Affiliation(s)
- Masahito Horiguchi
- Departments of Cell Biology and Medicine, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| | | | | |
Collapse
|
94
|
Callewaert B, Su CT, Van Damme T, Vlummens P, Malfait F, Vanakker O, Schulz B, Mac Neal M, Davis EC, Lee JGH, Salhi A, Unger S, Heimdal K, De Almeida S, Kornak U, Gaspar H, Bresson JL, Prescott K, Gosendi ME, Mansour S, Piérard GE, Madan-Khetarpal S, Sciurba FC, Symoens S, Coucke PJ, Van Maldergem L, Urban Z, De Paepe A. Comprehensive clinical and molecular analysis of 12 families with type 1 recessive cutis laxa. Hum Mutat 2012; 34:111-21. [PMID: 22829427 DOI: 10.1002/humu.22165] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 07/06/2012] [Indexed: 12/17/2022]
Abstract
Autosomal recessive cutis laxa type I (ARCL type I) is characterized by generalized cutis laxa with pulmonary emphysema and/or vascular complications. Rarely, mutations can be identified in FBLN4 or FBLN5. Recently, LTBP4 mutations have been implicated in a similar phenotype. Studying FBLN4, FBLN5, and LTBP4 in 12 families with ARCL type I, we found bi-allelic FBLN5 mutations in two probands, whereas nine probands harbored biallelic mutations in LTBP4. FBLN5 and LTBP4 mutations cause a very similar phenotype associated with severe pulmonary emphysema, in the absence of vascular tortuosity or aneurysms. Gastrointestinal and genitourinary tract involvement seems to be more severe in patients with LTBP4 mutations. Functional studies showed that most premature termination mutations in LTBP4 result in severely reduced mRNA and protein levels. This correlated with increased transforming growth factor-beta (TGFβ) activity. However, one mutation, c.4127dupC, escaped nonsense-mediated decay. The corresponding mutant protein (p.Arg1377Alafs(*) 27) showed reduced colocalization with fibronectin, leading to an abnormal morphology of microfibrils in fibroblast cultures, while retaining normal TGFβ activity. We conclude that LTBP4 mutations cause disease through both loss of function and gain of function mechanisms.
Collapse
Affiliation(s)
- Bert Callewaert
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Fischer B, Dimopoulou A, Egerer J, Gardeitchik T, Kidd A, Jost D, Kayserili H, Alanay Y, Tantcheva-Poor I, Mangold E, Daumer-Haas C, Phadke S, Peirano RI, Heusel J, Desphande C, Gupta N, Nanda A, Felix E, Berry-Kravis E, Kabra M, Wevers RA, van Maldergem L, Mundlos S, Morava E, Kornak U. Further characterization of ATP6V0A2-related autosomal recessive cutis laxa. Hum Genet 2012; 131:1761-73. [PMID: 22773132 DOI: 10.1007/s00439-012-1197-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 06/21/2012] [Indexed: 12/17/2022]
Abstract
Autosomal recessive cutis laxa (ARCL) syndromes are phenotypically overlapping, but genetically heterogeneous disorders. Mutations in the ATP6V0A2 gene were found to underlie both, autosomal recessive cutis laxa type 2 (ARCL2), Debré type, and wrinkly skin syndrome (WSS). The ATP6V0A2 gene encodes the a2 subunit of the V-type H(+)-ATPase, playing a role in proton translocation, and possibly also in membrane fusion. Here, we describe a highly variable phenotype in 13 patients with ARCL2, including the oldest affected individual described so far, who showed strikingly progressive dysmorphic features and heterotopic calcifications. In these individuals we identified 17 ATP6V0A2 mutations, 14 of which are novel. Furthermore, we demonstrate a localization of ATP6V0A2 at the Golgi-apparatus and a loss of the mutated ATP6V0A2 protein in patients' dermal fibroblasts. Investigation of brefeldin A-induced Golgi collapse in dermal fibroblasts as well as in HeLa cells deficient for ATP6V0A2 revealed a delay, which was absent in cells deficient for the ARCL-associated proteins GORAB or PYCR1. Furthermore, fibroblasts from patients with ATP6V0A2 mutations displayed elevated TGF-β signalling and increased TGF-β1 levels in the supernatant. Our current findings expand the genetic and phenotypic spectrum and suggest that, besides the known glycosylation defect, alterations in trafficking and signalling processes are potential key events in the pathogenesis of ATP6V0A2-related ARCL.
Collapse
Affiliation(s)
- Björn Fischer
- Institut fuer Medizinische Genetik und Humangenetik, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Mukherjee A, Larson EA, Carlos AS, Belknap JK, Rotwein P, Klein RF. Congenic mice provide in vivo evidence for a genetic locus that modulates intrinsic transforming growth factor β1-mediated signaling and bone acquisition. J Bone Miner Res 2012; 27:1345-56. [PMID: 22407846 PMCID: PMC4729561 DOI: 10.1002/jbmr.1590] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Osteoporosis, the most common skeletal disorder, is characterized by low bone mineral density (BMD) and an increased risk of fragility fractures. BMD is the best clinical predictor of future osteoporotic fracture risk, but is a complex trait controlled by multiple environmental and genetic determinants with individually modest effects. Quantitative trait locus (QTL) mapping is a powerful method for identifying chromosomal regions encompassing genes involved in shaping complex phenotypes, such as BMD. Here we have applied QTL analysis to male and female genetically-heterogeneous F(2) mice derived from a cross between C57BL/6 and DBA/2 strains, and have identified 11 loci contributing to femoral BMD. Further analysis of a QTL on mouse chromosome 7 following the generation of reciprocal congenic strains has allowed us to determine that the high BMD trait, which tracks with the DBA/2 chromosome and exerts equivalent effects on male and female mice, is manifested by enhanced osteogenic differentiation of mesenchymal stem cells (MSCs) in vitro and by increased growth of metatarsal bones in short-term primary culture. An insertion/deletion DNA polymorphism in Ltbp4 exon 12 that causes the in-frame removal of 12 codons in the DBA/2-derived gene maps within 0.6 Mb of the marker most tightly linked to the QTL. LTBP4, one of four paralogous mouse proteins that modify the bioavailability of the transforming growth factor β (TGF-β) family of growth factors, is expressed in differentiating MSC-derived osteoblasts and in long bones, and reduced responsiveness to TGF-β1 is observed in MSCs of mice homozygous for the DBA/2 chromosome 7. Taken together, our results identify a potential genetic and biochemical relationship between decreased TGF-β1-mediated signaling and enhanced femoral BMD that may be regulated by a variant LTBP4 molecule.
Collapse
Affiliation(s)
- Aditi Mukherjee
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR, United States
| | - Emily A. Larson
- Bone and Mineral Research Unit, Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Amy S. Carlos
- Bone and Mineral Research Unit, Department of Medicine, Oregon Health & Science University, Portland, OR, United States
| | - John K. Belknap
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Portland Veterans Affairs Medical Center, Portland, OR, United States
| | - Peter Rotwein
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, OR, United States
| | - Robert F. Klein
- Bone and Mineral Research Unit, Department of Medicine, Oregon Health & Science University, Portland, OR, United States
- Portland Veterans Affairs Medical Center, Portland, OR, United States
| |
Collapse
|
97
|
Doyle JJ, Gerber EE, Dietz HC. Matrix-dependent perturbation of TGFβ signaling and disease. FEBS Lett 2012; 586:2003-15. [PMID: 22641039 PMCID: PMC3426037 DOI: 10.1016/j.febslet.2012.05.027] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 05/13/2012] [Accepted: 05/15/2012] [Indexed: 02/07/2023]
Abstract
Transforming growth factor beta (TGFβ) is a multipotent cytokine that is sequestered in the extracellular matrix (ECM) through interactions with a number of ECM proteins. The ECM serves to concentrate latent TGFβ at sites of intended function, to influence the bioavailability and/or function of TGFβ activators, and perhaps to regulate the intrinsic performance of cell surface effectors of TGFβ signal propagation. The downstream consequences of TGFβ signaling cascades in turn provide feedback modulation of the ECM. This review covers recent examples of how genetic mutations in constituents of the ECM or TGFβ signaling cascade result in altered ECM homeostasis, cellular performance and ultimately disease, with an emphasis on emerging therapeutic strategies that seek to capitalize on this refined mechanistic understanding.
Collapse
|
98
|
Berk DR, Bentley DD, Bayliss SJ, Lind A, Urban Z. Cutis laxa: A review. J Am Acad Dermatol 2012; 66:842.e1-17. [DOI: 10.1016/j.jaad.2011.01.004] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 12/05/2010] [Accepted: 01/03/2011] [Indexed: 12/17/2022]
|
99
|
Abstract
Latent transforming growth factor beta (TGF-β) binding proteins (LTBPs) are large extracellular glycoproteins structurally similar to fibrillins. They perform intricate and important roles in the extracellular matrix (ECM) and perturbations of their function manifest as a wide range of diseases. LTBPs are major regulators of TGF-β bioavailability and action. In addition, LTBPs interact with other ECM proteins-from cytokines to large multi-factorial aggregates like microfibrils and elastic fibers, affecting their genesis, structure, and performance. In the present article, we review recent advancements in the field and relate the complex roles of LTBP in development and homeostasis.
Collapse
Affiliation(s)
- Vesna Todorovic
- Department of Cell Biology, NYU Langone Medical Center, New York, New York 10016, USA.
| | | |
Collapse
|
100
|
Leppäranta O, Sens C, Salmenkivi K, Kinnula VL, Keski-Oja J, Myllärniemi M, Koli K. Regulation of TGF-β storage and activation in the human idiopathic pulmonary fibrosis lung. Cell Tissue Res 2012; 348:491-503. [PMID: 22434388 DOI: 10.1007/s00441-012-1385-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 02/21/2012] [Indexed: 11/30/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease of unknown cause. The pathogenesis of the disease is characterized by fibroblast accumulation and excessive transforming growth factor-β (TGF-β) activation. Although TGF-β activation is a complex process involving various protein interactions, little is known of the specific routes of TGF-β storage and activation in human lung. Here, we have systematically analyzed the expression of specific proteins involved in extracellular matrix targeting and activation of TGF-β. Latent TGF-β-binding protein (LTBP)-1 was found to be significantly upregulated in IPF patient lungs. LTBP-1 expression was especially high in the fibroblastic foci, in which P-Smad2 immunoreactivity, indicative of TGF-β signaling activity, was less prominent. In cultured primary lung fibroblasts and epithelial cells, short-interfering-RNA-mediated downregulation of LTBP-1 resulted in either increased or decreased TGF-β signaling activity, respectively, suggesting that LTBP-1-mediated TGF-β activation is dependent on the cellular context in the lung. Furthermore, LTBP-1 was shown to colocalize with fibronectin, fibrillin-1 and fibrillin-2 proteins in the IPF lung. Fibrillin-2, a developmental gene expressed only in blood vessels in normal adult lung, was found specifically upregulated in IPF fibroblastic foci. The TGF-β-activating integrin β8 subunit was expressed at low levels in both control and IPF lungs. Alterations in extracellular matrix composition, such as high levels of the TGF-β storage protein LTBP-1 and the re-appearance of fibrillin-2, probably modulate TGF-β availability and activation in different pulmonary compartments in the fibrotic lung.
Collapse
Affiliation(s)
- Outi Leppäranta
- Department of Medicine, Division of Pulmonary Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | | | | | | | | | | | | |
Collapse
|