A Novel Regulatory Mechanism of the BMP Signaling Pathway

Total Page:16

File Type:pdf, Size:1020Kb

A Novel Regulatory Mechanism of the BMP Signaling Pathway A novel regulatory mechanism of the BMP signaling pathway Jose Maria Amich Manero under the direction of Prof. Vicki Rosen Dr. Jonathan Lowery Harvard School of Dental Medicine Research Science Institute July 30, 2013 Abstract The BMP signaling pathway is a pivotal morphogenetic signal involved in a wide spectrum of cellular processes. The fact that the number of ligands far exceeds the number of receptors, and how a limited canonical pathway can accomplish pleiotropic effects demonstrate that the regulation of this pathway is, at present, poorly understood. In this study, we propose N-linked glycosylation as a specific regulatory mechanism of the BMP type 2 receptors (ACVR2A, ACVR2B and BMPR2). Computational screening for glycosylated asparagine residues in BMPR2 reveals three putative sites, which we show to be glycosylated by means of site-directed mutagenesis. Furthermore, we demonstrate that BMPR2 glycosylation is essential for ligand binding but that glycosylation of ACVR2A prevents binding. Collectively, our findings provide the first mechanistic insight into the regulation of the BMP signaling pathway through glycosylation of BMP type 2 receptors. Summary Numerous organismic processes such as embryonic development and bone growth are controlled by a cell regulatory mechanism known as the bone morphogenetic protein (BMP) pathway. This pathway is activated when a signaling protein binds to the membrane receptor, transmitting in turn an order to the nucleus. In an attempt to shed light on BMP pathway activation, we focused on receptor-bound sugar chains in view of their protein-specific signa- ture. To study the role of individual sugar chains, we systematically blocked their function until we were able to pinpoint three key sites. We also assessed if these sugars affected the receptor's ligand-binding ability, and found that they promoted binding to the receptor in some cases and prevented binding in others. Thus, our findings provide the first mechanistic explanation for the BMP pathway regulation at a receptor-specific level. 1 Introduction 1.1 The TGF-β superfamily The bone morphogenetic protein (BMP) signaling pathway plays a key role in the regulation of many cellular processes, such as embryo and cell growth, differentiation and apoptosis [1]. The BMP pathway has been observed to be necessary for the maintenance of vascular and bone homeostasis, leading to severe disease when mutations occur in members of the pathway [2]. The BMP ligands belong to the TGF-β superfamily, featuring many other proteins such as growth and differentiation factors (GDFs) that regulate cartilage and skeletal development [3], Activins (Acts) and inhibins (Inhs) that regulate pituitary hormone secretion [4], the M¨ullerianinhibiting substance (MIS) that determines sex during embryonic development [5] and bone morphogenetic proteins (BMPs) that regulate vertebrate development [6]. Receptor extracellular ligand binding is necessary for the activation of the BMP signaling pathway. Mechanistically, the initial activation of the pathway is achieved via the binding of transforming growth factor β-like (TGF-β-like) ligands to the Activin/TGF-β and BMP receptors, leading to cellular response through the regulation of target genes. 1.2 Structure and function of BMPs BMPs have recently been under extensive study due to their implications in human disease. BMPs were originally described by Urist in 1965 [7], who observed that extracts of this protein had the ability to induce osteogenesis after intramuscular implantation. Since then, BMPs have been reported to contribute to a wide range of processes, such as cartilage development, osteogenesis and oocyte development [8]. BMPs bind to the extracellular domain (ECD) of the type II receptor (BMPR2, ACVR2A and ACVR2B) [9], which dimerizes with another receptor of its type. The dimeric type II 1 Figure 1: The activation of the BMP signaling pathway occurs when a BMP ligand triggers dimerization of type 1 (ALK2, 3, 6) and type 2 (BMPR2, ACVR2A, ACVR2B) receptors, leading to phosphorylation of SMAD1/5/8 and translocating to the nucleus together with SMAD4 for transcription of genes containing the SMAD binding elements. receptors will phosphorylate the GS box of the type I receptors (ALK2, ALK3 and ALK6) [9], inducing the type I receptor to phosphorylate the C-terminus of several receptor-activated (RA) SMAD proteins (SMAD1, SMAD5 and SMAD8) [10]. The RA-SMAD complex will then bind to SMAD4 and translocate as a RA-SMAD/SMAD4 complex to the nucleus, facilitating the transcription of genes by binding to SMAD binding elements (SBEs) (Figure 1) [11]. The number of characterized BMP ligands outweighs the number of known receptors, giving rise to a competitive and synergistic receptor activation network. Competition for the bone morphogenetic protein 2 (BMP2) has been identified as a potent regulator of the canon- ical SMAD pathway (SMAD1/5/8) [12]. Several studies have reported that affinity of BMP2 to type 2 receptors ACVR2B and ACVR2A is higher than to BMPR2 [13, 14]. As ACVR2A and ACVR2B also bind to Activin, eliciting the receptor-activated SMAD2/3 canonical 2 pathway, which is often opposite in effect to SMAD1/5/8, it is possible that BMPR2 has a regulatory role in SMAD signaling based on BMP and Activin competition. However, many of these studies have been performed using bacterially-expressed proteins, which might lead to artifactual differences based on altered posttranslational modifications. One such possible modification is N-linked glycosylation, which does not occur in bacteria. 1.3 The role of glycosylation in BMP binding kinetics Glycosylation is a protein posttranslational modification based on the addition of polysac- charides to key residues, playing a role in protein structure and function [15]. The importance of glycosylation in determining a protein's properties has been shown to be critical in a num- ber of cases, such as hormones and cytokines [15]. In the case of secreted signaling proteins, glycosylation can alter their conformation and binding kinetics, and thus control pathway activation [15]. N-linked glycolysation occurs when a monosaccharide, N-acetylglucosamine, attaches to a nitrogen molecule in an asparagine residue. In eukaryotes, glycosylation takes place in the endoplasmic reticulum (ER) lumen, for a further cleavage to the cellular membrane [16]. While N-linked glycosylation has been well characterized in a number of proteins, its effect on the type 2 BMP receptor is still to be clarified. Kang et al [17] showed that a non- N-glycosylated pro-VEGF region significantly reduced VEGF secretion in Saccharomyces cerevisiae, demonstrating that N-linked glycosylation has a bearing in protein stability. In another study, Zheng et al [18] found that thermal stability and susceptibility to degradation by papain were dependent on glycosylation status, although protein secondary and tertiary structure were unchanged. 3 1.4 Research approach In this study, we aim to explore the structural and functional repercussions of aberrant or null BMP type 2 receptor glycosylation in BMP2 binding. Using several BMPR2 models containing single, double and triple substitutions in glycosylated asparagine residues, we assess BMP2 binding to BMPR2, as well as to the other type II receptors (ACVR2A and ACVR2B) to further understand the specific role of glycosylation at the individual receptor level. We hope our findings will shed light on BMP type 2 receptor function, enabling a clearer understanding of their complex and highly specific regulatory mechanism. 2 Methods 2.1 hBMPR2 plasmid constructions Prior to the start of RSI, several plasmids were generated in order to study the effects of site- directed mutagenesis on hBMPR2 function. A shuttle plasmid encoding the human BMPR2 was subcloned into the carbenicillin-resistant pcDNA3.1/V5-His-TOPO expression vector (Invitrogen), which appends C-terminal V5 and His epitope tags in-frame, via standard PCR using GoTag HotStart DNA Polymerase (Promega). The obtained plasmids were hBMPR2, hBMPR2- N55Q, hBMPR2 -N110Q, hBMPR2- N126Q, hBMPR2- N55Q/ N110Q, hBMPR2- N55Q/ N110Q, hBMPR2- N55Q/ N126Q, hBMPR2- N110Q/ N126Q and hBMPR2- N55Q/ N110Q/ N126Q. The primers used for plasmid sequencing plasmids are shown in the ap- pendix (Appendix A.1). 2.2 W20 transfections Mutant species of hBMPR2 were obtained via the transfection of W20 cells with plasmids containing the single, double and triple mutant hBMPR2 sequences. Before transfecting 4 the cells, Escherichia coli bacteria were transformed in order to propagate the plasmids. To effectuate the transformation, we firstly transfered 50µl of bacteria and added 1µg of plasmid DNA. To permeabilize the bacterial membrane, the culture was heated up to 42oC during 20 seconds and selection was performed in carbenicillin-containing media. The culture was further incubated at 32oC with shaking, and 100µl of the bacteria were then transferred into a new agar plate for further growth overnight. After incubation, the cultures were pelleted at 13000rpm and the plasmids isolated following the QIAprep Spin MiniPrep kit (QIAGEN) instructions. The transfection was performed using the X-tremeGENE 9 DNA Transfection Reagent (Roche) on W20 cells, previously passaged in 10% DMEM. To perform the transfection, the X-tremeGENE 9 DNA Transfection Reagent was vortexed and 3µl of the reagent was added to 100µl of serum-free medium (to a concentration of 3:1 reagent per DNA ratio). At this point, 1µg of DNA was added to the 100µl of diluted reagent, and the solution was incubated for 15 minutes at room temperature. As the W20 cells were grown in a 25cm2 dish, 300µl of the diluted reagent was further added to the cell culture vessels in a dropwise manner without removal of the media. Lysis of the cells was performed using RIPA buffer 72 hours after transfection, where the proteins were retrieved in the supernatant phase and subsequently prepared for western blotting. 2.3 Immunoblotting Western blots were performed on lysates from transfected W20 cells and BMPR2-ECD/Fc chimeras. Proteins were resolved on Novex Tris Glycine gels (Invitrogen) and transferred to Amersham Hybond ECL nitrocellulose membranes (GE Healthcare).
Recommended publications
  • Characterization of Protein Kinase C Alpha Deficiency in a Mouse Model
    Aus der Medizinischen Klinik mit Schwerpunkt Infektiologie und Pneumologie der Charité – Universitätsmedizin Berlin Eingereicht über das Institut für Veterinär-Physiologie des Fachbereichs Veterinärmedizin der Freien Universität Berlin Characterization of Protein Kinase C Alpha Deficiency in a Mouse Model Inaugural-Dissertation zur Erlangung des Doctor of Philosophy (Ph.D.) an der Freien Universität Berlin vorgelegt von Elena Ariane Noe Tierärztin aus Düsseldorf Berlin 2016 Journal-Nr.: 3878 Gedruckt mit Genehmigung des Fachbereichs Veterinärmedizin der Freien Universität Berlin Dekan: Univ.-Prof. Dr. Jürgen Zentek Erster Gutachter: Prof. Dr. Dr. Petra Reinhold Zweiter Gutachter: Univ.-Prof. Dr. Martin Witzenrath Dritter Gutachter: Univ.-Prof. Dr. Christa Thöne-Reineke Deskriptoren (nach CAB-Thesaurus): Mice; animal models; protein kinase C (MeSH); pulmonary artery; hypertension; blood pressure, vasoconstriction; esophageal sphincter, lower (MeSH); respiratory system; smooth muscle; esophageal achalasia (MeSH) Tag der Promotion: 14.07.2016 Contents Contents ................................................................................................................................... V List of Abbreviations ............................................................................................................... VII 1 Introduction ................................................................................................................. 1 1.1 Protein Kinase C (PKC) and its Role in Smooth Muscle Contraction ........................
    [Show full text]
  • BMPR2 Mutations in Pulmonary Arterial Hypertension with Congenital Heart Disease
    Copyright #ERS Journals Ltd 2004 Eur Respir J 2004; 24: 371–374 European Respiratory Journal DOI: 10.1183/09031936.04.00018604 ISSN 0903-1936 Printed in UK – all rights reserved BMPR2 mutations in pulmonary arterial hypertension with congenital heart disease K.E. Roberts*, J.J. McElroy#, W.P.K. Wong*, E. Yen*, A. Widlitz}, R.J. Barst}, J.A. Knowles#,z,§, J.H. Morse* # } BMPR2 mutations in pulmonary arterial hypertension with congenital heart disease. Depts ofz *Medicine, Psychiatry, Pediatrics, K.E. Roberts, J.J. McElroy, W.P.K. Wong, E. Yen, A. Widlitz, R.J. Barst, J.A. Knowles, and the Columbia Genome Center, Columbia University College of Physicians and Surgeons, J.H. Morse. #ERS Journals Ltd 2004. § ABSTRACT: The aim of the present study was to determine if patients with both and the New York State Psychiatric Institute, New York, NY, USA. pulmonary arterial hypertension (PAH), due to pulmonary vascular obstructive disease, and congenital heart defects (CHD), have mutations in the gene encoding bone Correspondence: J.H. Morse, Dept of Medi- morphogenetic protein receptor (BMPR)-2. cine, Columbia University College of Physi- The BMPR2 gene was screened in two cohorts: 40 adults and 66 children with PAH/ cians and Surgeons, New York, NY, USA. CHD. CHDs were patent ductus arteriosus, atrial and ventricular septal defects, partial Fax: 1 2123054943 anomalous pulmonary venous return, transposition of the great arteries, atrioventicular E-mail: [email protected] canal, and rare lesions with systemic-to-pulmonary shunts. Six novel missense BMPR2 mutations were found in three out of four adults with Keywords: Bone morphogenetic protein receptor 2 mutations complete type C atrioventricular canals and in three children.
    [Show full text]
  • The BMP Receptor 2 in Pulmonary Arterial Hypertension: When and Where the Animal Model Matches the Patient
    cells Article The BMP Receptor 2 in Pulmonary Arterial Hypertension: When and Where the Animal Model Matches the Patient 1, 2, 1 Chris Happé y, Kondababu Kurakula y, Xiao-Qing Sun , Denielli da Silva Goncalves Bos 1 , Nina Rol 1, Christophe Guignabert 3,4 , Ly Tu 3,4 , Ingrid Schalij 1, Karien C. Wiesmeijer 2, Olga Tura-Ceide 5,6,7, Anton Vonk Noordegraaf 1, Frances S. de Man 1, Harm Jan Bogaard 1 and Marie-José Goumans 2,* 1 Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pulmonology, Amsterdam Cardiovascular Sciences, 1081 HV Amsterdam, The Netherlands; [email protected] (C.H.); [email protected] (X.-Q.S.); [email protected] (D.d.S.G.B.); [email protected] (N.R.); [email protected] (I.S.); [email protected] (A.V.N.); [email protected] (F.S.d.M.); [email protected] (H.J.B.) 2 Laboratory for Cardiovascular Cell Biology, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; [email protected] (K.K.); [email protected] (K.C.W.) 3 INSERM UMR_S 999, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France; [email protected] (C.G.); [email protected] (L.T.) 4 Université Paris-Saclay, School of Medicine, 94270 Le Kremlin-Bicêtre, France 5 Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; [email protected] 6 Biomedical Research Networking center on Respiratory diseases (CIBERES), 28029 Madrid, Spain 7 Department of Pulmonary Medicine, Dr.
    [Show full text]
  • The Role of Genetics Mutations in Genes ACVR1, BMPR1A, BMPR1B, BMPR2, BMP4 in Stone Man Syndrome
    Asadi S and Aranian MR, J Hematol Hemother 5: 008 Journal of Hematology & Hemotherapy Review Article The Role of Genetics Mutations in Genes ACVR1, BMPR1A, BMPR1B, BMPR2, BMP4 in Stone Man Syndrome Asadi S* and Aranian MR Division of Medical Genetics and Molecular Pathology Research, Harvard University, Boston Children’s Hospital, Iran Abstract *Corresponding author: Shahin Asadi, Division of Medical Genetics and Molecular Pathology Research, Harvard University, Boston Children’s Hospital, Iran, Tel: +98 Fibrodysplasia Ossificans Progressiva (FOP) is a severely dis- 9379923364; E-mail: [email protected] abling heritable disorder of connective tissue characterized by con- genital malformations of the great toes and progressive heterotopic Received Date: February 7, 2020 ossification that forms qualitatively normal bone in characteristic ex- Accepted Date: February 17, 2020 traskeletal sites. Classic FOP is caused by a recurrent activating mu- tation (617G>A; R206H) in the gene ACVR1 (ALK2) encoding Activin Published Date: February 24, 2020 A receptor type I/Activin-like kinase 2, a bone morphogenetic protein (BMP) type I receptor. Atypical FOP patients also have heterozygous Citation: Asadi S, Aranian MR (2020) The Role of Genetics Mutations in Genes ACVR1, BMPR1A, BMPR1B, BMPR2, BMP4 in Stone Man Syndrome. J Hematol ACVR1 missense mutations in conserved amino acids. Hemother 5: 008. Keywords: ACVR1; BMPR1A; BMPR1B; BMPR2; BMP4; Genetics Copyright: © 2020 Asadi S, et al. This is an open-access article distributed under the mutations, Stone man syndrome terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source Overview of Stone Man Syndrome are credited.
    [Show full text]
  • Somatic Ephrin Receptor Mutations Are Associated with Metastasis in Primary Colorectal Cancer Running Title
    Author Manuscript Published OnlineFirst on January 20, 2017; DOI: 10.1158/0008-5472.CAN-16-1921 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Uvyr)Thvp ru v rpr hv h r hpvhrq vu rhhv v vh py rphy phpr Svt vyr) 6u ) " #$ % & '$ $( $)*# $ ,$$ - ./ 0 1 " , $'2 / ' - , $$03$, 4, 5$ $ 6 7$ % & 8 8$ 9 6ssvyvhv) . 8 : .2 $ 5 1 $ ; $ ;# < %. 8 : ., , $ ;# $ < -: . $, 8 $ . . 8 ; $ ;# /. 8 : . $; $ ;# < 0= $ 1 2 . , >2,, $ ?&, $ 2 . & $ , $@ ABA%B, $ < 4: . $ $ $ ; $ ;# < 6: .2 $ 5 1 $ .@ $ $ $ = $ ; $ ;# < Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2017 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 20, 2017; DOI: 10.1158/0008-5472.CAN-16-1921 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. & 8 * $$ < 9& $8 C 8 < 8$ D< < 8syvp s vr rC & . $@ $, $ ', $E $ . $ 8$:7'@ < % Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2017 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 20, 2017; DOI: 10.1158/0008-5472.CAN-16-1921 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 6i hpC& 8 . . $ $ >? $ <& . # $# . $ $ .464A6 22)2 . $. E<& # $ >?. $ . ). $ . ) . $$ 222 2 < $ $ . @ . <& 8$. $ ., ::)$$@,. ) $ ),@$$<F .8 222 . , $) ,::)$$ ), $E <& 8# # $8 , < - Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2017 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 20, 2017; DOI: 10.1158/0008-5472.CAN-16-1921 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. D qpv #$ .$$) $ .. 8 >&5?>?<& . 8@ #$ $#$ # $ 8$ .8 .
    [Show full text]
  • BMPR1A Is Necessary for Chondrogenesis and Osteogenesis
    © 2020. Published by The Company of Biologists Ltd | Journal of Cell Science (2020) 133, jcs246934. doi:10.1242/jcs.246934 RESEARCH ARTICLE BMPR1A is necessary for chondrogenesis and osteogenesis, whereas BMPR1B prevents hypertrophic differentiation Tanja Mang1,2, Kerstin Kleinschmidt-Doerr1, Frank Ploeger3, Andreas Schoenemann4, Sven Lindemann1 and Anne Gigout1,* ABSTRACT essential for osteogenesis and bone formation during this process BMP2 stimulates bone formation and signals preferably through BMP (Bandyopadhyay et al., 2006; McBride et al., 2014; Yang et al., 2013). – receptor (BMPR) 1A, whereas GDF5 is a cartilage inducer and signals Similarly, during bone fracture healing where a similar mechanism – preferably through BMPR1B. Consequently, BMPR1A and BMPR1B are takes place conditional deletion of Bmp2 in mesenchymal believed to be involved in bone and cartilage formation, respectively. progenitors or osteoprogenitors prevents fracture healing (Mi et al., However, their function is not yet fully clarified. In this study, GDF5 2013; Tsuji et al., 2006). In vitro, BMP2 provokes an induction of mutants with a decreased affinity for BMPR1A were generated. These alkaline phosphatase (ALP) activity, osteocalcin expression and matrix mutants, and wild-type GDF5 and BMP2, were tested for their ability to mineralization in pluripotent mesenchymal progenitor cells (Cheng induce dimerization of BMPR1A or BMPR1B with BMPR2, and for their et al., 2003), and also stimulates chondrogenesis or adipogenesis (Date chondrogenic, hypertrophic and osteogenic properties in chondrocytes, et al., 2004). Finally, BMP2 has been shown to promote bone repair in in the multipotent mesenchymal precursor cell line C3H10T1/2 and the animal models (Kleinschmidt et al., 2013; Wulsten et al., 2011) and in human osteosarcoma cell line Saos-2.
    [Show full text]
  • Inhibition of ERK 1/2 Kinases Prevents Tendon Matrix Breakdown Ulrich Blache1,2,3, Stefania L
    www.nature.com/scientificreports OPEN Inhibition of ERK 1/2 kinases prevents tendon matrix breakdown Ulrich Blache1,2,3, Stefania L. Wunderli1,2,3, Amro A. Hussien1,2, Tino Stauber1,2, Gabriel Flückiger1,2, Maja Bollhalder1,2, Barbara Niederöst1,2, Sandro F. Fucentese1 & Jess G. Snedeker1,2* Tendon extracellular matrix (ECM) mechanical unloading results in tissue degradation and breakdown, with niche-dependent cellular stress directing proteolytic degradation of tendon. Here, we show that the extracellular-signal regulated kinase (ERK) pathway is central in tendon degradation of load-deprived tissue explants. We show that ERK 1/2 are highly phosphorylated in mechanically unloaded tendon fascicles in a vascular niche-dependent manner. Pharmacological inhibition of ERK 1/2 abolishes the induction of ECM catabolic gene expression (MMPs) and fully prevents loss of mechanical properties. Moreover, ERK 1/2 inhibition in unloaded tendon fascicles suppresses features of pathological tissue remodeling such as collagen type 3 matrix switch and the induction of the pro-fbrotic cytokine interleukin 11. This work demonstrates ERK signaling as a central checkpoint to trigger tendon matrix degradation and remodeling using load-deprived tissue explants. Tendon is a musculoskeletal tissue that transmits muscle force to bone. To accomplish its biomechanical function, tendon tissues adopt a specialized extracellular matrix (ECM) structure1. Te load-bearing tendon compart- ment consists of highly aligned collagen-rich fascicles that are interspersed with tendon stromal cells. Tendon is a mechanosensitive tissue whereby physiological mechanical loading is vital for maintaining tendon archi- tecture and homeostasis2. Mechanical unloading of the tissue, for instance following tendon rupture or more localized micro trauma, leads to proteolytic breakdown of the tissue with severe deterioration of both structural and mechanical properties3–5.
    [Show full text]
  • Cardiopulmonary and Neurologic Dysfunctions in Fibrodysplasia Ossificans Progressiva
    biomedicines Review Cardiopulmonary and Neurologic Dysfunctions in Fibrodysplasia Ossificans Progressiva Fatima Khan 1, Xiaobing Yu 2 and Edward C. Hsiao 3,* 1 Department of Medical Sciences, Frank H. Netter MD School of Medicine at Quinnipiac University, North Haven, CT 06518, USA; [email protected] 2 Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA 94143, USA; [email protected] 3 Department of Medicine, Division of Endocrinology and Metabolism, the Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA 94143, USA * Correspondence: [email protected]; Tel.: +1-(415)-476-9732 Abstract: Fibrodysplasia Ossificans Progressiva (FOP) is an ultra-rare but debilitating disorder char- acterized by spontaneous, progressive, and irreversible heterotopic ossifications (HO) at extraskeletal sites. FOP is caused by gain-of-function mutations in the Activin receptor Ia/Activin-like kinase 2 gene (Acvr1/Alk2), with increased receptor sensitivity to bone morphogenetic proteins (BMPs) and a neoceptor response to Activin A. There is extensive literature on the skeletal phenotypes in FOP, but a much more limited understanding of non-skeletal manifestations of this disease. Emerging evidence reveals important cardiopulmonary and neurologic dysfunctions in FOP including thoracic insufficiency syndrome, pulmonary hypertension, conduction abnormalities, neuropathic pain, and demyelination of the central nervous system (CNS). Here, we review the recent research and discuss unanswered questions regarding the cardiopulmonary and neurologic phenotypes in FOP. Keywords: Fibrodysplasia Ossificans Progressiva (FOP); cardiac conduction abnormalities; ACVR1; Citation: Khan, F.; Yu, X.; Hsiao, E.C. Cardiopulmonary and Neurologic neuropathic pain; cardiac dysfunction; neurological dysfunction Dysfunctions in Fibrodysplasia Ossificans Progressiva.
    [Show full text]
  • BMPR2 Germline Mutations in Pulmonary Hypertension Associated with Fenfluramine Derivatives
    Copyright #ERS Journals Ltd 2002 Eur Respir J 2002; 20: 518–523 European Respiratory Journal DOI: 10.1183/09031936.02.01762002 ISSN 0903-1936 Printed in UK – all rights reserved BMPR2 germline mutations in pulmonary hypertension associated with fenfluramine derivatives M. Humbert*, Z. Deng#,ƒ, G. Simonneau*, R.J. Barst},§, O. Sitbon*, M. Wolfz, N. Cuervo#, K.J. Moore#, S.E. Hodge#,ƒ,**, J.A. Knowles#,ƒ, J.H. Morse} z BMPR2 germline mutations in pulmonary hypertension associated with fenfluramine *Service de Pneumologie, and He´mato- derivatives. M. Humbert, Z. Deng, G. Simonneau, R.J. Barst, O. Sitbon, M. Wolf, logie, Hoˆpital Antoine Be´cle`re, Clamart, # France, and the Depts of }Medicine, N. Cuervo, K.J. Moore, S.E. Hodge, J.A. Knowles, J.H. Morse. ERS Journals Ltd § # 2002. Pediatrics, and Psychiatry, Columbia University College of Physicians and ABSTRACT: This study investigated whether patients developing pulmonary arterial Surgeons, ƒNew York State Psychiatric hypertension (PAH) after exposure to the appetite suppressants fenfluramine and Institute, and **Division of Biostatis- dexfenfluramine have mutations in the bone morphogenetic protein receptor 2 tics, Columbia University School of (BMPR2) gene, as reported in primary pulmonary hypertension. Public Health, New York, NY, USA. BMPR2 was examined for mutations in 33 unrelated patients with sporadic PAH, and in two sisters with PAH, all of whom had taken fenfluramine derivatives, as well as Correspondence: J.H. Morse, Dept in 130 normal controls. The PAH patients also underwent cardiac catheterisation and of Medicine, Columbia Presbyterian body mass determinations. Medical Center, PH 8 East, Suite 101, 630 West 168th Street, New York, Three BMPR2 mutations predicting changes in the primary structure of the BMPR- NY 10032.
    [Show full text]
  • Signaling Receptors for TGF-B Family Members
    Downloaded from http://cshperspectives.cshlp.org/ on September 28, 2021 - Published by Cold Spring Harbor Laboratory Press Signaling Receptors for TGF-b Family Members Carl-Henrik Heldin1 and Aristidis Moustakas1,2 1Ludwig Institute for Cancer Research Ltd., Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden 2Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23 Uppsala, Sweden Correspondence: [email protected] Transforming growth factor b (TGF-b) family members signal via heterotetrameric complexes of type I and type II dual specificity kinase receptors. The activation and stability of the receptors are controlled by posttranslational modifications, such as phosphorylation, ubiq- uitylation, sumoylation, and neddylation, as well as by interaction with other proteins at the cell surface and in the cytoplasm. Activation of TGF-b receptors induces signaling via formation of Smad complexes that are translocated to the nucleus where they act as tran- scription factors, as well as via non-Smad pathways, including the Erk1/2, JNK and p38 MAP kinase pathways, and the Src tyrosine kinase, phosphatidylinositol 30-kinase, and Rho GTPases. he transforming growth factor b (TGF-b) embryonic development and in the regulation Tfamily of cytokine genes has 33 human of tissue homeostasis, through their abilities to members, encoding TGF-b isoforms, bone regulate cell proliferation, migration, and differ- morphogenetic proteins (BMPs), growth and entiation. Perturbation of signaling by TGF-b differentiation factors (GDFs), activins, inhib- family members is often seen in different dis- ins, nodal, and anti-Mu¨llerian hormone (AMH) eases, including malignancies, inflammatory (Derynck and Miyazono 2008; Moustakas and conditions, and fibrotic conditions.
    [Show full text]
  • Genetic Analyses in a Cohort of 191 Pulmonary Arterial Hypertension
    Yang et al. Respiratory Research (2018) 19:87 https://doi.org/10.1186/s12931-018-0789-9 RESEARCH Open Access Genetic analyses in a cohort of 191 pulmonary arterial hypertension patients Hang Yang1†, Qixian Zeng2†, Yanyun Ma1†, Bingyang Liu2, Qianlong Chen1, Wenke Li1, Changming Xiong2,3* and Zhou Zhou1,3* Abstract Background: Pulmonary arterial hypertension (PAH) is a progressive and fatal disorder associated with high pulmonary artery pressure. Genetic testing enables early diagnosis and offers an opportunity for family screening. To identify genetic mutations and help make a precise diagnosis, we performed genetic testing in 191 probands with PAH and tried to analyze the genotype-phenotype correlation. Methods: Initially, PAH samples (n = 119) were submitted to BMPR2 screening using Sanger sequencing. Later, we developed a PAH panel test to identify causal mutations in 13 genes related to PAH and tried to call BMPR2 copy number variations (CNVs) with the panel data. Multiplex ligation-dependent probe amplification (MLPA) was used to search for CNVs in BMPR2, ACVRL1 and ENG.Notably,EIF2AK4 gene was also involved in the panel, which allowed to distinguish pulmonary veno-occlusive disease (PVOD)/pulmonary capillary hemangiomatosis (PCH) patients from idiopathic PAH (IPAH). Characteristics of patients were compared using t test for continuous variables. Results: Pathogenic BMPR2 mutations were detected most frequently in 32 (17.9%) IPAH and 5 (41.7%) heritable PAH (HPAH) patients by sequencing, and 12 BMPR2 CNVs called from the panel data were all successfully confirmed by MLPA analysis. In addition, homozygous or compound heterozygous EIF2AK4 mutations were identified in 6 patients, who should be corrected to a diagnosis of PVOD/PCH.
    [Show full text]
  • BMPR2 Inhibits Activin- and BMP-Signaling Via Wild Type ALK2
    bioRxiv preprint doi: https://doi.org/10.1101/222406; this version posted November 22, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 BMPR2 inhibits activin- and BMP-signaling via wild type ALK2 2 3 Oddrun Elise Olsen1,2, Meenu Sankar3, Samah Elsaadi1, Hanne Hella1, Glenn Buene1, Sagar 4 Ramesh Darvekar1, Kristine Misund1,2, Takenobu Katagiri4 and Toril Holien1,2* 5 6 (1) Department of Clinical and Molecular Medicine, NTNU – Norwegian University of 7 Science and Technology, Trondheim, Norway. 8 (2) Clinic of Medicine, St. Olav’s University Hospital, Trondheim, Norway. 9 (3) School of Bioscience, University of Skövde, Skövde, Sweden. 10 (4) Division of Pathophysiology, Research Center for Genomic Medicine, Saitama Medical 11 University, Hidaka-shi, Saitama 350-1241, Japan. 12 13 * Corresponding author 14 E-mail: [email protected] 15 16 17 Running title: BMPR2 inhibits ALK2 activity 18 19 Key words: Bone Morphogenetic Protein, BMPR2, ACVR2A, ACVR2B, Activin, ACVR1 1 bioRxiv preprint doi: https://doi.org/10.1101/222406; this version posted November 22, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 Summary Statement 2 The activation of SMAD1/5/8 via endogenous wild type ALK2 by activin A, activin B, and 3 certain BMPs was enhanced when BMPR2 levels were knocked down. 4 5 6 Abstract 7 Activin A is a member of the TGF-β superfamily and activates the transcription factors 8 SMAD2/3 through the ALK4 type 1 receptor.
    [Show full text]