1
|
Amioka N, Franklin MK, Kukida M, Zhu L, Moorleghen JJ, Howatt DA, Katsumata Y, Mullick AE, Yanagita M, Martinez-Irizarry MM, Sandoval RM, Dunn KW, Sawada H, Daugherty A, Lu HS. Renal Proximal Tubule Cell-Specific Megalin Deletion Does Not Affect Atherosclerosis But Induces Tubulointerstitial Nephritis in Mice Fed a Western Diet. Arterioscler Thromb Vasc Biol 2025; 45:74-89. [PMID: 39569521 PMCID: PMC11668626 DOI: 10.1161/atvbaha.124.321366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Pharmacological inhibition of megalin (also known as LRP2 [low-density lipoprotein receptor-related protein-2]) attenuates atherosclerosis in hypercholesterolemic mice. Since megalin is abundant in renal proximal tubule cells (PTCs), the purpose of this study was to determine whether PTC-specific deletion of megalin reduces hypercholesterolemia-induced atherosclerosis in mice. METHODS Female Lrp2 f/f mice were bred with male Ndrg1-Cre ERT2 +/0 mice to develop PTC-LRP2 +/+ and PTC-LRP2 -/- littermates. To study atherosclerosis, all mice were bred to an LDL (low-density lipoprotein) receptor -/- background and fed a Western diet to induce atherosclerosis. RESULTS PTC-specific megalin deletion did not attenuate atherosclerosis in LDL receptor -/- mice in either sex. Serendipitously, we discovered that PTC-specific megalin deletion led to interstitial infiltration of CD68+ cells and tubular atrophy. The pathology was only evident in male PTC-LRP2 -/- mice fed a Western diet but not in mice fed a normal laboratory diet. Renal pathologies were also observed in male PTC-LRP2 -/- mice in an LDL receptor +/+ background fed the same Western diet, demonstrating that the renal pathologies were dependent on diet and not on hypercholesterolemia. In contrast, female PTC-LRP2 -/- mice had no apparent renal pathologies. In vivo multiphoton microscopy demonstrated that PTC-specific megalin deletion dramatically diminished ALB (albumin) accumulation in PTCs within 10 days of Western diet feeding. RNA-sequencing analyses demonstrated the upregulation of inflammation-related pathways in the kidney. CONCLUSIONS PTC-specific megalin deletion does not affect atherosclerosis but leads to tubulointerstitial nephritis in mice fed a Western diet, with severe pathologies in male mice.
Collapse
MESH Headings
- Animals
- Low Density Lipoprotein Receptor-Related Protein-2/genetics
- Low Density Lipoprotein Receptor-Related Protein-2/metabolism
- Low Density Lipoprotein Receptor-Related Protein-2/deficiency
- Diet, Western/adverse effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Female
- Male
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/prevention & control
- Mice, Knockout
- Disease Models, Animal
- Nephritis, Interstitial/genetics
- Nephritis, Interstitial/pathology
- Nephritis, Interstitial/metabolism
- Mice
- Mice, Inbred C57BL
- Hypercholesterolemia/genetics
- Hypercholesterolemia/complications
- Hypercholesterolemia/metabolism
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Plaque, Atherosclerotic
- CD68 Molecule
Collapse
Affiliation(s)
- Naofumi Amioka
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Michael K. Franklin
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Masayoshi Kukida
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Liyuan Zhu
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Jessica J. Moorleghen
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Deborah A. Howatt
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Biostatistics, University of Kentucky, Lexington, Kentucky, USA
| | | | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | | | - Ruben M. Sandoval
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Kenneth W. Dunn
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center and Saha Aortic Center, University of Kentucky, Lexington, Kentucky, USA
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
2
|
Amioka N, Franklin MK, Kukida M, Zhu L, Moorleghen JJ, Howatt DA, Katsumata Y, Mullick AE, Yanagita M, Martinez-Irizarry MM, Sandoval RM, Dunn KW, Sawada H, Daugherty A, Lu HS. Renal Proximal Tubule Cell-specific Megalin Deletion Does Not Affect Atherosclerosis But Induces Tubulointerstitial Nephritis in Mice Fed Western Diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.592234. [PMID: 38798535 PMCID: PMC11118422 DOI: 10.1101/2024.05.11.592234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background Pharmacological inhibition of megalin (also known as low-density lipoprotein receptor-related protein 2: LRP2) attenuates atherosclerosis in hypercholesterolemic mice. Since megalin is abundant in renal proximal tubule cells (PTCs), the purpose of this study was to determine whether PTC-specific deletion of megalin reduces hypercholesterolemia-induced atherosclerosis in mice. Methods Female Lrp2 f/f mice were bred with male Ndrg1-Cre ERT2 +/0 mice to develop PTC-LRP2 +/+ and -/- littermates. To study atherosclerosis, all mice were bred to an LDL receptor -/- background and fed a Western diet to induce atherosclerosis. Results PTC-specific megalin deletion did not attenuate atherosclerosis in LDL receptor -/- mice in either sex. Serendipitously, we discovered that PTC-specific megalin deletion led to interstitial infiltration of CD68+ cells and tubular atrophy. The pathology was only evident in male PTC-LRP2 -/- mice fed the Western diet, but not in mice fed a normal laboratory diet. Renal pathologies were also observed in male PTC-LRP2 -/- mice in an LDL receptor +/+ background fed the same Western diet, demonstrating that the renal pathologies were dependent on diet and not hypercholesterolemia. In contrast, female PTC-LRP2 -/- mice had no apparent renal pathologies. In vivo multiphoton microscopy demonstrated that PTC-specific megalin deletion dramatically diminished albumin accumulation in PTCs within 10 days of Western diet feeding. RNA sequencing analyses demonstrated the upregulation of inflammation-related pathways in kidney. Conclusions PTC-specific megalin deletion does not affect atherosclerosis, but leads to tubulointerstitial nephritis in mice fed Western diet, with severe pathologies in male mice.
Collapse
Affiliation(s)
- Naofumi Amioka
- Saha Cardiovascular Research Center and Saha Aortic Center
| | | | | | - Liyuan Zhu
- Saha Cardiovascular Research Center and Saha Aortic Center
| | | | | | | | | | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | | | - Ruben M. Sandoval
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Kenneth W. Dunn
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center and Saha Aortic Center
- Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
3
|
Gill E, Bamforth SD. Molecular Pathways and Animal Models of Truncus Arteriosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:853-865. [PMID: 38884754 DOI: 10.1007/978-3-031-44087-8_52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
In normal cardiovascular development in birds and mammals, the outflow tract of the heart is divided into two distinct channels to separate the oxygenated systemic blood flow from the deoxygenated pulmonary circulation. When the process of outflow tract septation fails, a single common outflow vessel persists resulting in a serious clinical condition known as persistent truncus arteriosus or common arterial trunk. In this chapter, we will review molecular pathways and the cells that are known to play a role in the formation and development of the outflow tract and how genetic manipulation of these pathways in animal models can result in common arterial trunk.
Collapse
Affiliation(s)
- Eleanor Gill
- Newcastle University Biosciences Institute, Newcastle, UK
| | | |
Collapse
|
4
|
LRP2 contributes to planar cell polarity-dependent coordination of motile cilia function. Cell Tissue Res 2023; 392:535-551. [PMID: 36764939 PMCID: PMC10172251 DOI: 10.1007/s00441-023-03757-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/04/2022] [Indexed: 02/12/2023]
Abstract
Motile cilia are protruding organelles on specialized epithelia that beat in a synchronous fashion to propel extracellular fluids. Coordination and orientation of cilia beating on individual cells and across tissues is a complex process dependent on planar cell polarity (PCP) signaling. Asymmetric sorting of PCP pathway components, essential to establish planar polarity, involves trafficking along the endocytic path, but the underlying regulatory processes remain incompletely understood. Here, we identified the endocytic receptor LRP2 as regulator of PCP component trafficking in ependyma, a multi-ciliated cell type that is involved in facilitating flow of the cerebrospinal fluid in the brain ventricular system. Lack of receptor expression in gene-targeted mice results in a failure to sort PCP core proteins to the anterior or posterior cell side and, consequently, in the inability to coordinate cilia arrangement and to aligned beating (loss of rotational and translational polarity). LRP2 deficiency coincides with a failure to sort NHERF1, a cytoplasmic LRP2 adaptor to the anterior cell side. As NHERF1 is essential to translocate PCP core protein Vangl2 to the plasma membrane, these data suggest a molecular mechanism whereby LRP2 interacts with PCP components through NHERF1 to control their asymmetric sorting along the endocytic path. Taken together, our findings identified the endocytic receptor LRP2 as a novel regulator of endosomal trafficking of PCP proteins, ensuring their asymmetric partition and establishment of translational and rotational planar cell polarity in the ependyma.
Collapse
|
5
|
Poelmann RE, Gittenberger-de Groot AC, Goerdajal C, Grewal N, De Bakker MAG, Richardson MK. Ventricular Septation and Outflow Tract Development in Crocodilians Result in Two Aortas with Bicuspid Semilunar Valves. J Cardiovasc Dev Dis 2021; 8:jcdd8100132. [PMID: 34677201 PMCID: PMC8537894 DOI: 10.3390/jcdd8100132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/14/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023] Open
Abstract
Background: The outflow tract of crocodilians resembles that of birds and mammals as ventricular septation is complete. The arterial anatomy, however, presents with a pulmonary trunk originating from the right ventricular cavum, and two aortas originating from either the right or left ventricular cavity. Mixing of blood in crocodilians cannot occur at the ventricular level as in other reptiles but instead takes place at the aortic root level by a shunt, the foramen of Panizza, the opening of which is guarded by two facing semilunar leaflets of both bicuspid aortic valves. Methods: Developmental stages of Alligator mississipiensis, Crocodilus niloticus and Caiman latirostris were studied histologically. Results and Conclusions: The outflow tract septation complex can be divided into two components. The aorto-pulmonary septum divides the pulmonary trunk from both aortas, whereas the interaortic septum divides the systemic from the visceral aorta. Neural crest cells are most likely involved in the formation of both components. Remodeling of the endocardial cushions and both septa results in the formation of bicuspid valves in all three arterial trunks. The foramen of Panizza originates intracardially as a channel in the septal endocardial cushion.
Collapse
Affiliation(s)
- Robert E. Poelmann
- Sylvius Laboratory, Department of Animal Sciences and Health, Institute of Biology, University of Leiden, Sylvi-usweg 72, 2333BE Leiden, The Netherlands; (C.G.); (M.A.G.D.B.); (M.K.R.)
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300RC Leiden, The Netherlands;
- Correspondence: ; Tel.: +31-652695875
| | | | - Charissa Goerdajal
- Sylvius Laboratory, Department of Animal Sciences and Health, Institute of Biology, University of Leiden, Sylvi-usweg 72, 2333BE Leiden, The Netherlands; (C.G.); (M.A.G.D.B.); (M.K.R.)
| | - Nimrat Grewal
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Albinusdreef 2, P.O. Box 9600, 2300RC Leiden, The Netherlands;
| | - Merijn A. G. De Bakker
- Sylvius Laboratory, Department of Animal Sciences and Health, Institute of Biology, University of Leiden, Sylvi-usweg 72, 2333BE Leiden, The Netherlands; (C.G.); (M.A.G.D.B.); (M.K.R.)
| | - Michael K. Richardson
- Sylvius Laboratory, Department of Animal Sciences and Health, Institute of Biology, University of Leiden, Sylvi-usweg 72, 2333BE Leiden, The Netherlands; (C.G.); (M.A.G.D.B.); (M.K.R.)
| |
Collapse
|
6
|
Mecklenburg N, Kowalczyk I, Witte F, Görne J, Laier A, Mamo TM, Gonschior H, Lehmann M, Richter M, Sporbert A, Purfürst B, Hübner N, Hammes A. Identification of disease-relevant modulators of the SHH pathway in the developing brain. Development 2021; 148:272000. [PMID: 34463328 DOI: 10.1242/dev.199307] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 07/19/2021] [Indexed: 12/13/2022]
Abstract
Pathogenic gene variants in humans that affect the sonic hedgehog (SHH) pathway lead to severe brain malformations with variable penetrance due to unknown modifier genes. To identify such modifiers, we established novel congenic mouse models. LRP2-deficient C57BL/6N mice suffer from heart outflow tract defects and holoprosencephaly caused by impaired SHH activity. These defects are fully rescued on a FVB/N background, indicating a strong influence of modifier genes. Applying comparative transcriptomics, we identified Pttg1 and Ulk4 as candidate modifiers upregulated in the rescue strain. Functional analyses showed that ULK4 and PTTG1, both microtubule-associated proteins, are positive regulators of SHH signaling, rendering the pathway more resilient to disturbances. In addition, we characterized ULK4 and PTTG1 as previously unidentified components of primary cilia in the neuroepithelium. The identification of genes that powerfully modulate the penetrance of genetic disturbances affecting the brain and heart is likely relevant to understanding the variability in human congenital disorders.
Collapse
Affiliation(s)
- Nora Mecklenburg
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Izabela Kowalczyk
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Franziska Witte
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Jessica Görne
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Alena Laier
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Tamrat M Mamo
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Hannes Gonschior
- Cellular Imaging, Light Microscopy, Leibniz-Research Institute for Molecular Pharmacology (FMP), 13125 Berlin, Germany
| | - Martin Lehmann
- Cellular Imaging, Light Microscopy, Leibniz-Research Institute for Molecular Pharmacology (FMP), 13125 Berlin, Germany
| | - Matthias Richter
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Bettina Purfürst
- Electron microscopy technology platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany.,Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany.,Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Annette Hammes
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| |
Collapse
|
7
|
Arrigo AB, Lin JHI. Endocytic Protein Defects in the Neural Crest Cell Lineage and Its Pathway Are Associated with Congenital Heart Defects. Int J Mol Sci 2021; 22:8816. [PMID: 34445520 PMCID: PMC8396181 DOI: 10.3390/ijms22168816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 02/02/2023] Open
Abstract
Endocytic trafficking is an under-appreciated pathway in cardiac development. Several genes related to endocytic trafficking have been uncovered in a mutagenic ENU screen, in which mutations led to congenital heart defects (CHDs). In this article, we review the relationship between these genes (including LRP1 and LRP2) and cardiac neural crest cells (CNCCs) during cardiac development. Mice with an ENU-induced Lrp1 mutation exhibit a spectrum of CHDs. Conditional deletion using a floxed Lrp1 allele with different Cre drivers showed that targeting neural crest cells with Wnt1-Cre expression replicated the full cardiac phenotypes of the ENU-induced Lrp1 mutation. In addition, LRP1 function in CNCCs is required for normal OFT lengthening and survival/expansion of the cushion mesenchyme, with other cell lineages along the NCC migratory path playing an additional role. Mice with an ENU-induced and targeted Lrp2 mutation demonstrated the cardiac phenotype of common arterial trunk (CAT). Although there is no impact on CNCCs in Lrp2 mutants, the loss of LRP2 results in the depletion of sonic hedgehog (SHH)-dependent cells in the second heart field. SHH is known to be crucial for CNCC survival and proliferation, which suggests LRP2 has a non-autonomous role in CNCCs. In this article, other endocytic trafficking proteins that are associated with CHDs that may play roles in the NCC pathway during development, such as AP1B1, AP2B1, FUZ, MYH10, and HECTD1, are reviewed.
Collapse
Affiliation(s)
- Angelo B. Arrigo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15224, USA;
| | - Jiuann-Huey Ivy Lin
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15224, USA;
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA
| |
Collapse
|
8
|
Peterson JC, Kelder TP, Goumans MJTH, Jongbloed MRM, DeRuiter MC. The Role of Cell Tracing and Fate Mapping Experiments in Cardiac Outflow Tract Development, New Opportunities through Emerging Technologies. J Cardiovasc Dev Dis 2021; 8:47. [PMID: 33925811 PMCID: PMC8146276 DOI: 10.3390/jcdd8050047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/18/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023] Open
Abstract
Whilst knowledge regarding the pathophysiology of congenital heart disease (CHDs) has advanced greatly in recent years, the underlying developmental processes affecting the cardiac outflow tract (OFT) such as bicuspid aortic valve, tetralogy of Fallot and transposition of the great arteries remain poorly understood. Common among CHDs affecting the OFT, is a large variation in disease phenotypes. Even though the different cell lineages contributing to OFT development have been studied for many decades, it remains challenging to relate cell lineage dynamics to the morphologic variation observed in OFT pathologies. We postulate that the variation observed in cellular contribution in these congenital heart diseases might be related to underlying cell lineage dynamics of which little is known. We believe this gap in knowledge is mainly the result of technical limitations in experimental methods used for cell lineage analysis. The aim of this review is to provide an overview of historical fate mapping and cell tracing techniques used to study OFT development and introduce emerging technologies which provide new opportunities that will aid our understanding of the cellular dynamics underlying OFT pathology.
Collapse
Affiliation(s)
- Joshua C. Peterson
- Department Anatomy & Embryology, Leiden University Medical Center, 2300RC Leiden, The Netherlands; (J.C.P.); (T.P.K.); (M.R.M.J.)
| | - Tim P. Kelder
- Department Anatomy & Embryology, Leiden University Medical Center, 2300RC Leiden, The Netherlands; (J.C.P.); (T.P.K.); (M.R.M.J.)
| | - Marie José T. H. Goumans
- Department Cellular and Chemical Biology, Leiden University Medical Center, 2300RC Leiden, The Netherlands;
| | - Monique R. M. Jongbloed
- Department Anatomy & Embryology, Leiden University Medical Center, 2300RC Leiden, The Netherlands; (J.C.P.); (T.P.K.); (M.R.M.J.)
- Department of Cardiology, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Marco C. DeRuiter
- Department Anatomy & Embryology, Leiden University Medical Center, 2300RC Leiden, The Netherlands; (J.C.P.); (T.P.K.); (M.R.M.J.)
| |
Collapse
|
9
|
Stefanovic S, Etchevers HC, Zaffran S. Outflow Tract Formation-Embryonic Origins of Conotruncal Congenital Heart Disease. J Cardiovasc Dev Dis 2021; 8:jcdd8040042. [PMID: 33918884 PMCID: PMC8069607 DOI: 10.3390/jcdd8040042] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 12/13/2022] Open
Abstract
Anomalies in the cardiac outflow tract (OFT) are among the most frequent congenital heart defects (CHDs). During embryogenesis, the cardiac OFT is a dynamic structure at the arterial pole of the heart. Heart tube elongation occurs by addition of cells from pharyngeal, splanchnic mesoderm to both ends. These progenitor cells, termed the second heart field (SHF), were first identified twenty years ago as essential to the growth of the forming heart tube and major contributors to the OFT. Perturbation of SHF development results in common forms of CHDs, including anomalies of the great arteries. OFT development also depends on paracrine interactions between multiple cell types, including myocardial, endocardial and neural crest lineages. In this publication, dedicated to Professor Andriana Gittenberger-De Groot and her contributions to the field of cardiac development and CHDs, we review some of her pioneering studies of OFT development with particular interest in the diverse origins of the many cell types that contribute to the OFT. We also discuss the clinical implications of selected key findings for our understanding of the etiology of CHDs and particularly OFT malformations.
Collapse
|
10
|
Schussler O, Gharibeh L, Mootoosamy P, Murith N, Tien V, Rougemont AL, Sologashvili T, Suuronen E, Lecarpentier Y, Ruel M. Cardiac Neural Crest Cells: Their Rhombomeric Specification, Migration, and Association with Heart and Great Vessel Anomalies. Cell Mol Neurobiol 2021; 41:403-429. [PMID: 32405705 PMCID: PMC11448677 DOI: 10.1007/s10571-020-00863-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Outflow tract abnormalities are the most frequent congenital heart defects. These are due to the absence or dysfunction of the two main cell types, i.e., neural crest cells and secondary heart field cells that migrate in opposite directions at the same stage of development. These cells directly govern aortic arch patterning and development, ascending aorta dilatation, semi-valvular and coronary artery development, aortopulmonary septation abnormalities, persistence of the ductus arteriosus, trunk and proximal pulmonary arteries, sub-valvular conal ventricular septal/rotational defects, and non-compaction of the left ventricle. In some cases, depending on the functional defects of these cells, additional malformations are found in the expected spatial migratory area of the cells, namely in the pharyngeal arch derivatives and cervico-facial structures. Associated non-cardiovascular anomalies are often underestimated, since the multipotency and functional alteration of these cells can result in the modification of multiple neural, epidermal, and cervical structures at different levels. In most cases, patients do not display the full phenotype of abnormalities, but congenital cardiac defects involving the ventricular outflow tract, ascending aorta, aortic arch and supra-aortic trunks should be considered as markers for possible impaired function of these cells. Neural crest cells should not be considered as a unique cell population but on the basis of their cervical rhombomere origins R3-R5 or R6-R7-R8 and specific migration patterns: R3-R4 towards arch II, R5-R6 arch III and R7-R8 arch IV and VI. A better understanding of their development may lead to the discovery of unknown associated abnormalities, thereby enabling potential improvements to be made to the therapeutic approach.
Collapse
Affiliation(s)
- Olivier Schussler
- Department of Cardiovascular Surgery Adult and Pediatric, Geneva University Hospital, Geneva, Switzerland.
- Cardiovascular Research Laboratory, Faculty of Medicine of the University of Geneva, Rue Michel Servet 1, 1211, Geneva 4, Switzerland.
| | - Lara Gharibeh
- Molecular Genetics and Cardiac Regeneration Laboratory, Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Parmeseeven Mootoosamy
- Department of Cardiovascular Surgery Adult and Pediatric, Geneva University Hospital, Geneva, Switzerland
| | - Nicolas Murith
- Department of Cardiovascular Surgery Adult and Pediatric, Geneva University Hospital, Geneva, Switzerland
| | - Vannary Tien
- Department of Pathology and Immunology, Faculty of Medicine of the University of Geneva, Geneva, Switzerland
| | | | - Tornike Sologashvili
- Department of Cardiovascular Surgery Adult and Pediatric, Geneva University Hospital, Geneva, Switzerland
| | - Erik Suuronen
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Cardiovascular Surgery, University of Ottawa Heart Institute and School of Epidemiology, Ottawa, ON, Canada
| | | | - Marc Ruel
- Department of Cardiovascular Surgery, University of Ottawa Heart Institute and School of Epidemiology, Ottawa, ON, Canada
| |
Collapse
|
11
|
Snider JC, Riley LA, Mallory NT, Bersi MR, Umbarkar P, Gautam R, Zhang Q, Mahadevan-Jansen A, Hatzopoulos AK, Maroteaux L, Lal H, Merryman WD. Targeting 5-HT 2B Receptor Signaling Prevents Border Zone Expansion and Improves Microstructural Remodeling After Myocardial Infarction. Circulation 2021; 143:1317-1330. [PMID: 33474971 PMCID: PMC8009826 DOI: 10.1161/circulationaha.120.051517] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 01/06/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Myocardial infarction (MI) induces an intense injury response that ultimately generates a collagen-dominated scar. Although required to prevent ventricular rupture, the fibrotic process is often sustained in a manner detrimental to optimal recovery. Cardiac myofibroblasts are the cells tasked with depositing and remodeling collagen and are a prime target to limit the fibrotic process after MI. Serotonin 2B receptor (5-HT2B) signaling has been shown to be harmful in a variety of cardiopulmonary pathologies and could play an important role in mediating scar formation after MI. METHODS We used 2 pharmacological antagonists to explore the effect of 5-HT2B inhibition on outcomes after MI and characterized the histological and microstructural changes involved in tissue remodeling. Inducible 5-HT2B ablation driven by Tcf21MCM and PostnMCM was used to evaluate resident cardiac fibroblast- and myofibroblast-specific contributions of 5-HT2B, respectively. RNA sequencing was used to motivate subsequent in vitro analyses to explore cardiac fibroblast phenotype. RESULTS 5-HT2B antagonism preserved cardiac structure and function by facilitating a less fibrotic scar, indicated by decreased scar thickness and decreased border zone area. 5-HT2B antagonism resulted in collagen fiber redistribution to thinner collagen fibers that were more anisotropic, enhancing left ventricular contractility, whereas fibrotic tissue stiffness was decreased, limiting the hypertrophic response of uninjured cardiomyocytes. Using a tamoxifen-inducible Cre, we ablated 5-HT2B from Tcf21-lineage resident cardiac fibroblasts and saw similar improvements to the pharmacological approach. Tamoxifen-inducible Cre-mediated ablation of 5-HT2B after onset of injury in Postn-lineage myofibroblasts also improved cardiac outcomes. RNA sequencing and subsequent in vitro analyses corroborate a decrease in fibroblast proliferation, migration, and remodeling capabilities through alterations in Dnajb4 expression and Src phosphorylation. CONCLUSIONS Together, our findings illustrate that 5-HT2B expression in either cardiac fibroblasts or activated myofibroblasts directly contributes to excessive scar formation, resulting in adverse remodeling and impaired cardiac function after MI.
Collapse
Affiliation(s)
- J. Caleb Snider
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232
| | - Lance A. Riley
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232
| | - Noah T. Mallory
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232
| | - Matthew R. Bersi
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232
| | - Prachi Umbarkar
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - Rekha Gautam
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232
| | - Qinkun Zhang
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294
| | | | - Antonis K. Hatzopoulos
- Division of Cardiovascular Medicine, Department of Medicine and Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Luc Maroteaux
- INSERM UMR-S 1270, 75005 Paris, France; Sorbonne Universités, 75005 Paris, France; Institut du Fer à Moulin, 75005 Paris, France
| | - Hind Lal
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294
| | - W. David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232
| |
Collapse
|
12
|
Kowalczyk I, Lee C, Schuster E, Hoeren J, Trivigno V, Riedel L, Görne J, Wallingford JB, Hammes A, Feistel K. Neural tube closure requires the endocytic receptor Lrp2 and its functional interaction with intracellular scaffolds. Development 2021; 148:dev195008. [PMID: 33500317 PMCID: PMC7860117 DOI: 10.1242/dev.195008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/11/2020] [Indexed: 12/20/2022]
Abstract
Pathogenic mutations in the endocytic receptor LRP2 in humans are associated with severe neural tube closure defects (NTDs) such as anencephaly and spina bifida. Here, we have combined analysis of neural tube closure in mouse and in the African Clawed Frog Xenopus laevis to elucidate the etiology of Lrp2-related NTDs. Lrp2 loss of function impaired neuroepithelial morphogenesis, culminating in NTDs that impeded anterior neural plate folding and neural tube closure in both model organisms. Loss of Lrp2 severely affected apical constriction as well as proper localization of the core planar cell polarity (PCP) protein Vangl2, demonstrating a highly conserved role of the receptor in these processes, which are essential for neural tube formation. In addition, we identified a novel functional interaction of Lrp2 with the intracellular adaptor proteins Shroom3 and Gipc1 in the developing forebrain. Our data suggest that, during neurulation, motifs within the intracellular domain of Lrp2 function as a hub that orchestrates endocytic membrane removal for efficient apical constriction, as well as PCP component trafficking in a temporospatial manner.
Collapse
Affiliation(s)
- Izabela Kowalczyk
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - Chanjae Lee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Elisabeth Schuster
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Josefine Hoeren
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Valentina Trivigno
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| | - Levin Riedel
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - Jessica Görne
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - John B Wallingford
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Annette Hammes
- Disorders of the Nervous System, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Robert Rössle Strasse 10, 13125 Berlin, Germany
| | - Kerstin Feistel
- University of Hohenheim, Institute of Biology, Department of Zoology, Garbenstrasse 30, 70599 Stuttgart, Germany
| |
Collapse
|
13
|
Basualto-Alarcón C, Llanos P, García-Rivas G, Troncoso MF, Lagos D, Barrientos G, Estrada M. Classic and Novel Sex Hormone Binding Globulin Effects on the Cardiovascular System in Men. Int J Endocrinol 2021; 2021:5527973. [PMID: 34335746 PMCID: PMC8318754 DOI: 10.1155/2021/5527973] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
In men, 70% of circulating testosterone binds with high affinity to plasma sex hormone binding globulin (SHBG), which determines its bioavailability in their target cells. In recent years, a growing body of evidence has shown that circulating SHBG not only is a passive carrier for steroid hormones but also actively regulates testosterone signaling through putative plasma membrane receptors and by local expression of androgen-binding proteins apparently to reach local elevated testosterone concentrations in specific androgen target tissues. Circulating SHBG levels are influenced by metabolic and hormonal factors, and they are reduced in obesity and insulin resistance, suggesting that SHBG may have a broader clinical utility in assessing the risk for cardiovascular diseases. Importantly, plasma SHBG levels are strongly correlated with testosterone concentrations, and in men, low testosterone levels are associated with an adverse cardiometabolic profile. Although obesity and insulin resistance are associated with an increased incidence of cardiovascular disease, whether they lead to abnormal expression of circulating SHBG or its interaction with androgen signaling remains to be elucidated. SHBG is produced mainly in the liver, but it can also be expressed in several tissues including the brain, fat tissue, and myocardium. Expression of SHBG is controlled by peroxisome proliferator-activated receptor γ (PPARγ) and AMP-activated protein kinase (AMPK). AMPK/PPAR interaction is critical to regulate hepatocyte nuclear factor-4 (HNF4), a prerequisite for SHBG upregulation. In cardiomyocytes, testosterone activates AMPK and PPARs. Therefore, the description of local expression of cardiac SHBG and its circulating levels may shed new light to explain physiological and adverse cardiometabolic roles of androgens in different tissues. According to emerging clinical evidence, here, we will discuss the potential mechanisms with cardioprotective effects and SHBG levels to be used as an early metabolic and cardiovascular biomarker in men.
Collapse
Affiliation(s)
- Carla Basualto-Alarcón
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique 5951537, Chile
- Departamento de Anatomía y Medicina Legal, Facultad de Medicina, Universidad de Chile, Santiago 8389100, Chile
| | - Paola Llanos
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Gerardo García-Rivas
- Tecnológico de Monterrey, Hospital Zambrano Hellion, TecSalud, Centro de Medicina Funcional, San Pedro Garza García, Nuevo León 66278, Mexico
| | - Mayarling Francisca Troncoso
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8389100, Chile
| | - Daniel Lagos
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8389100, Chile
| | - Genaro Barrientos
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8389100, Chile
| | - Manuel Estrada
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8389100, Chile
| |
Collapse
|
14
|
Christ A, Marczenke M, Willnow TE. LRP2 controls sonic hedgehog-dependent differentiation of cardiac progenitor cells during outflow tract formation. Hum Mol Genet 2020; 29:3183-3196. [PMID: 32901292 PMCID: PMC7689296 DOI: 10.1093/hmg/ddaa200] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Conotruncal malformations are a major cause of congenital heart defects in newborn infants. Recently, genetic screens in humans and in mouse models have identified mutations in LRP2, a multi-ligand receptor, as a novel cause of a common arterial trunk, a severe form of outflow tract (OFT) defect. Yet, the underlying mechanism why the morphogen receptor LRP2 is essential for OFT development remained unexplained. Studying LRP2-deficient mouse models, we now show that LRP2 is expressed in the cardiac progenitor niche of the anterior second heart field (SHF) that contributes to the elongation of the OFT during separation into aorta and pulmonary trunk. Loss of LRP2 in mutant mice results in the depletion of a pool of sonic hedgehog-dependent progenitor cells in the anterior SHF due to premature differentiation into cardiomyocytes as they migrate into the OFT myocardium. Depletion of this cardiac progenitor cell pool results in aberrant shortening of the OFT, the likely cause of CAT formation in affected mice. Our findings identified the molecular mechanism whereby LRP2 controls the maintenance of progenitor cell fate in the anterior SHF essential for OFT separation, and why receptor dysfunction is a novel cause of conotruncal malformation.
Collapse
Affiliation(s)
- Annabel Christ
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Maike Marczenke
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| |
Collapse
|
15
|
Theis JL, Vogler G, Missinato MA, Li X, Nielsen T, Zeng XXI, Martinez-Fernandez A, Walls SM, Kervadec A, Kezos JN, Birker K, Evans JM, O'Byrne MM, Fogarty ZC, Terzic A, Grossfeld P, Ocorr K, Nelson TJ, Olson TM, Colas AR, Bodmer R. Patient-specific genomics and cross-species functional analysis implicate LRP2 in hypoplastic left heart syndrome. eLife 2020; 9:e59554. [PMID: 33006316 PMCID: PMC7581429 DOI: 10.7554/elife.59554] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022] Open
Abstract
Congenital heart diseases (CHDs), including hypoplastic left heart syndrome (HLHS), are genetically complex and poorly understood. Here, a multidisciplinary platform was established to functionally evaluate novel CHD gene candidates, based on whole-genome and iPSC RNA sequencing of a HLHS family-trio. Filtering for rare variants and altered expression in proband iPSCs prioritized 10 candidates. siRNA/RNAi-mediated knockdown in healthy human iPSC-derived cardiomyocytes (hiPSC-CM) and in developing Drosophila and zebrafish hearts revealed that LDL receptor-related protein LRP2 is required for cardiomyocyte proliferation and differentiation. Consistent with hypoplastic heart defects, compared to patents the proband's iPSC-CMs exhibited reduced proliferation. Interestingly, rare, predicted-damaging LRP2 variants were enriched in a HLHS cohort; however, understanding their contribution to HLHS requires further investigation. Collectively, we have established a multi-species high-throughput platform to rapidly evaluate candidate genes and their interactions during heart development, which are crucial first steps toward deciphering oligogenic underpinnings of CHDs, including hypoplastic left hearts.
Collapse
Affiliation(s)
- Jeanne L Theis
- Cardiovascular Genetics Research LaboratoryRochesterUnited States
| | - Georg Vogler
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Maria A Missinato
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Xing Li
- Division of Biomedical Statistics and Informatics, Mayo ClinicRochesterUnited States
| | - Tanja Nielsen
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
- Doctoral Degrees and Habilitations, Department of Biology, Chemistry, and Pharmacy, Freie Universität BerlinBerlinGermany
| | - Xin-Xin I Zeng
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | | | - Stanley M Walls
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Anaïs Kervadec
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - James N Kezos
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Katja Birker
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Jared M Evans
- Division of Biomedical Statistics and Informatics, Mayo ClinicRochesterUnited States
| | - Megan M O'Byrne
- Division of Biomedical Statistics and Informatics, Mayo ClinicRochesterUnited States
| | - Zachary C Fogarty
- Division of Biomedical Statistics and Informatics, Mayo ClinicRochesterUnited States
| | - André Terzic
- Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Department of Molecular and Pharmacology and Experimental Therapeutics, Mayo ClinicLa JollaUnited States
- Center for Regenerative Medicine, Mayo ClinicRochesterUnited States
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo ClinicRochesterUnited States
| | - Paul Grossfeld
- University of California San Diego, Rady’s HospitalSan DiegoUnited States
- Division of General Internal Medicine, Mayo ClinicRochesterUnited States
| | - Karen Ocorr
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Timothy J Nelson
- Department of Molecular and Pharmacology and Experimental Therapeutics, Mayo ClinicLa JollaUnited States
- Center for Regenerative Medicine, Mayo ClinicRochesterUnited States
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo ClinicRochesterUnited States
| | - Timothy M Olson
- Department of Cardiovascular Medicine, Mayo ClinicRochesterUnited States
- Department of Molecular and Pharmacology and Experimental Therapeutics, Mayo ClinicLa JollaUnited States
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, Mayo ClinicRochesterUnited States
| | - Alexandre R Colas
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| | - Rolf Bodmer
- Development, Aging and Regeneration, Sanford Burnham Prebys Medical Discovery InstituteLa JollaUnited States
| |
Collapse
|
16
|
Johnson AL, Schneider JE, Mohun TJ, Williams T, Bhattacharya S, Henderson DJ, Phillips HM, Bamforth SD. Early Embryonic Expression of AP-2α Is Critical for Cardiovascular Development. J Cardiovasc Dev Dis 2020; 7:jcdd7030027. [PMID: 32717817 PMCID: PMC7570199 DOI: 10.3390/jcdd7030027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 12/17/2022] Open
Abstract
Congenital cardiovascular malformation is a common birth defect incorporating abnormalities of the outflow tract and aortic arch arteries, and mice deficient in the transcription factor AP-2α (Tcfap2a) present with complex defects affecting these structures. AP-2α is expressed in the pharyngeal surface ectoderm and neural crest at mid-embryogenesis in the mouse, but the precise tissue compartment in which AP-2α is required for cardiovascular development has not been identified. In this study we describe the fully penetrant AP-2α deficient cardiovascular phenotype on a C57Bl/6J genetic background and show that this is associated with increased apoptosis in the pharyngeal ectoderm. Neural crest cell migration into the pharyngeal arches was not affected. Cre-expressing transgenic mice were used in conjunction with an AP-2α conditional allele to examine the effect of deleting AP-2α from the pharyngeal surface ectoderm and the neural crest, either individually or in combination, as well as the second heart field. This, surprisingly, was unable to fully recapitulate the global AP-2α deficient cardiovascular phenotype. The outflow tract and arch artery phenotype was, however, recapitulated through early embryonic Cre-mediated recombination. These findings indicate that AP-2α has a complex influence on cardiovascular development either being required very early in embryogenesis and/or having a redundant function in many tissue layers.
Collapse
Affiliation(s)
- Amy-Leigh Johnson
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
| | | | | | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Anshutz Medical Campus, Aurora, CO 80045, USA;
| | - Shoumo Bhattacharya
- Department of Cardiovascular Medicine, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford OX3 7BN, UK;
| | - Deborah J. Henderson
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
| | - Helen M. Phillips
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
| | - Simon D. Bamforth
- Newcastle University Biosciences Institute, Centre for Life, Newcastle NE1 3BZ, UK; (A.-L.J.); (D.J.H.); (H.M.P.)
- Correspondence: ; Tel.: +44-191-241-8764
| |
Collapse
|
17
|
Kukida M, Sawada H, Daugherty A, Lu HS. Megalin: A bridge connecting kidney, the renin-angiotensin system, and atherosclerosis. Pharmacol Res 2020; 151:104537. [PMID: 31707037 PMCID: PMC6980733 DOI: 10.1016/j.phrs.2019.104537] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/05/2019] [Indexed: 01/21/2023]
Abstract
Megalin is a member of the low-density lipoprotein receptor superfamily. It has been recognized as an endocytic receptor for a large spectrum of ligands. As a consequence, megalin regulates homeostasis of many molecules and affects multiple physiological and pathophysiological functions. The renin-angiotensin system is a hormonal system. A number of studies have reported contributions of the renin-angiotensin system to atherosclerosis. There is evolving evidence that megalin is a regulator of the renin-angiotensin system, and contributes to atherosclerosis. This brief review provides contemporary insights into effects of megalin on renal functions, the renin-angiotensin system, and atherosclerosis.
Collapse
Affiliation(s)
- Masayoshi Kukida
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Hisashi Sawada
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Alan Daugherty
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Hong S Lu
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
18
|
Sasai N, Toriyama M, Kondo T. Hedgehog Signal and Genetic Disorders. Front Genet 2019; 10:1103. [PMID: 31781166 PMCID: PMC6856222 DOI: 10.3389/fgene.2019.01103] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
The hedgehog (Hh) family comprises sonic hedgehog (Shh), Indian hedgehog (Ihh), and desert hedgehog (Dhh), which are versatile signaling molecules involved in a wide spectrum of biological events including cell differentiation, proliferation, and survival; establishment of the vertebrate body plan; and aging. These molecules play critical roles from embryogenesis to adult stages; therefore, alterations such as abnormal expression or mutations of the genes involved and their downstream factors cause a variety of genetic disorders at different stages. The Hh family involves many signaling mediators and functions through complex mechanisms, and achieving a comprehensive understanding of the entire signaling system is challenging. This review discusses the signaling mediators of the Hh pathway and their functions at the cellular and organismal levels. We first focus on the roles of Hh signaling mediators in signal transduction at the cellular level and the networks formed by these factors. Then, we analyze the spatiotemporal pattern of expression of Hh pathway molecules in tissues and organs, and describe the phenotypes of mutant mice. Finally, we discuss the genetic disorders caused by malfunction of Hh signaling-related molecules in humans.
Collapse
Affiliation(s)
- Noriaki Sasai
- Developmental Biomedical Science, Division of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Michinori Toriyama
- Systems Neurobiology and Medicine, Division of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
19
|
Sandireddy R, Cibi DM, Gupta P, Singh A, Tee N, Uemura A, Epstein JA, Singh MK. Semaphorin 3E/PlexinD1 signaling is required for cardiac ventricular compaction. JCI Insight 2019; 4:125908. [PMID: 31434798 DOI: 10.1172/jci.insight.125908] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 05/01/2019] [Indexed: 01/10/2023] Open
Abstract
Left ventricular noncompaction (LVNC) is one of the most common forms of genetic cardiomyopathy characterized by excessive trabeculation and impaired myocardial compaction during fetal development. Patients with LVNC are at higher risk of developing left/right ventricular failure or both. Although the key regulators for cardiac chamber development are well studied, the role of semaphorin (Sema)/plexin signaling in this process remains poorly understood. In this article, we demonstrate that genetic deletion of Plxnd1, a class-3 Sema receptor in endothelial cells, leads to severe cardiac chamber defects. They were characterized by excessive trabeculation and noncompaction similar to patients with LVNC. Loss of Plxnd1 results in decreased expression of extracellular matrix proteolytic genes, leading to excessive deposition of cardiac jelly. We demonstrate that Plxnd1 deficiency is associated with an increase in Notch1 expression and its downstream target genes. In addition, inhibition of the Notch signaling pathway partially rescues the excessive trabeculation and noncompaction phenotype present in Plxnd1 mutants. Furthermore, we demonstrate that Semaphorin 3E (Sema3E), one of PlexinD1's known ligands, is expressed in the developing heart and is required for myocardial compaction. Collectively, our study uncovers what we believe to be a previously undescribed role of the Sema3E/PlexinD1 signaling pathway in myocardial trabeculation and the compaction process.
Collapse
Affiliation(s)
- Reddemma Sandireddy
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | - Dasan Mary Cibi
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | - Priyanka Gupta
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | - Anamika Singh
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore
| | - Nicole Tee
- National Heart Research Institute Singapore, National Heart Center Singapore, Singapore
| | - Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Japan
| | - Jonathan A Epstein
- Penn Cardiovascular Institute, Department of Medicine, Department of Cell and Developmental Biology, and Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Manvendra K Singh
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School Singapore, Singapore.,National Heart Research Institute Singapore, National Heart Center Singapore, Singapore
| |
Collapse
|
20
|
Wang JJ, Liu HX, Song L, Li HR, Yang YP, Zhang T, Jing Y. Isl-1 positive pharyngeal mesenchyme subpopulation and its role in the separation and remodeling of the aortic sac in embryonic mouse heart. Dev Dyn 2019; 248:771-783. [PMID: 31175693 DOI: 10.1002/dvdy.68] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 05/26/2019] [Accepted: 05/31/2019] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Second heart field cells and neural crest cells have been reported to participate in the morphogenesis of the pharyngeal arch arteries (PAAs); however, how the PAAs grow out and are separated from the aortic sac into left and right sections is unknown. RESULTS An Isl-1 positive pharyngeal mesenchyme protrusion in the aortic sac ventrally extends and fuses with the aortic sac wall to form a midsagittal septum that divides the aortic sac. The aortic sac division separates the left and right PAAs to form independent arteries. The midsagittal septum dividing the aortic sac has a different expression pattern from the aortic-pulmonary (AP) septum in which Isl-1 positive cells are absent. At 11 days post-conception (dpc) in a mouse embryo, the Isl-1 positive mesenchyme protrusion appears as a heart-shaped structure, in which subpopulations with Isl-1+ Tbx3+ and Isl-1+ Nkx2.5+ cells are included. CONCLUSIONS The aortic sac is a dynamic structure that is continuously divided during the migration from the pharyngeal mesenchyme to the pericardial cavity. The separation of the aortic sac is not complete until the AP septum divides the aortic sac into the ascending aorta and pulmonary trunk. Moreover, the midsagittal septum and the AP septum are distinct structures.
Collapse
Affiliation(s)
- Jing-Jing Wang
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hui-Xia Liu
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Song
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hai-Rong Li
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yan-Ping Yang
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tao Zhang
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ya Jing
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
21
|
Poelmann RE, Gittenberger-de Groot AC. Development and evolution of the metazoan heart. Dev Dyn 2019; 248:634-656. [PMID: 31063648 PMCID: PMC6767493 DOI: 10.1002/dvdy.45] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 12/19/2022] Open
Abstract
The mechanisms of the evolution and development of the heart in metazoans are highlighted, starting with the evolutionary origin of the contractile cell, supposedly the precursor of cardiomyocytes. The last eukaryotic common ancestor is likely a combination of several cellular organisms containing their specific metabolic pathways and genetic signaling networks. During evolution, these tool kits diversified. Shared parts of these conserved tool kits act in the development and functioning of pumping hearts and open or closed circulations in such diverse species as arthropods, mollusks, and chordates. The genetic tool kits became more complex by gene duplications, addition of epigenetic modifications, influence of environmental factors, incorporation of viral genomes, cardiac changes necessitated by air‐breathing, and many others. We evaluate mechanisms involved in mollusks in the formation of three separate hearts and in arthropods in the formation of a tubular heart. A tubular heart is also present in embryonic stages of chordates, providing the septated four‐chambered heart, in birds and mammals passing through stages with first and second heart fields. The four‐chambered heart permits the formation of high‐pressure systemic and low‐pressure pulmonary circulation in birds and mammals, allowing for high metabolic rates and maintenance of body temperature. Crocodiles also have a (nearly) separated circulation, but their resting temperature conforms with the environment. We argue that endothermic ancestors lost the capacity to elevate their body temperature during evolution, resulting in ectothermic modern crocodilians. Finally, a clinically relevant paragraph reviews the occurrence of congenital cardiac malformations in humans as derailments of signaling pathways during embryonic development. The cardiac regulatory toolkit contains many factors including epigenetic, genetic, viral, hemodynamic, and environmental factors, but also transcriptional activators, repressors, duplicated genes, redundancies and dose‐dependancies. Numerous toolkits regulate mechanisms including cell‐cell interactions, EMT, mitosis patterns, cell migration and differentiation and left/right sidedness involved in the development of endocardial cushions, looping, septum complexes, pharyngeal arch arteries, chamber and valve formation and conduction system. Evolutionary development of the yolk sac circulation likely preceded the advent of endothermy in amniotes. Parallel evolutionary traits regulate the development of contractile pumps in various taxa often in conjunction with the gut, lungs and excretory organs.
Collapse
Affiliation(s)
- Robert E Poelmann
- Institute of Biology, Department of Animal Sciences and Health, Leiden University, Leiden, The Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
22
|
Liu Y, Chen H, Shou W. Potential Common Pathogenic Pathways for the Left Ventricular Noncompaction Cardiomyopathy (LVNC). Pediatr Cardiol 2018; 39:1099-1106. [PMID: 29766225 PMCID: PMC6093786 DOI: 10.1007/s00246-018-1882-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 04/24/2018] [Indexed: 01/01/2023]
Abstract
Ventricular trabeculation and compaction are two essential morphogenetic events for generating a functionally competent ventricular wall. A significant reduction in trabeculation is usually associated with hypoplastic wall and ventricular compact zone deficiencies, which commonly leads to embryonic heart failure and early embryonic lethality. In contrast, the arrest of ventricular wall compaction (noncompaction) is believed to be causative to the left ventricular noncompaction (LVNC), a genetically heterogeneous disorder and the third most common cardiomyopathy among pediatric patients. After critically reviewing recent findings from genetically engineered mouse models, we suggest a model which proposes that defects in myofibrillogenesis and polarization in trabecular cardiomyocytes underly the common pathogenic mechanism for ventricular noncompaction.
Collapse
Affiliation(s)
- Ying Liu
- Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hanying Chen
- Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Weinian Shou
- Riley Heart Research Center, Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
23
|
Xu J, Wu Q, Wang L, Han J, Pei Y, Zhi W, Liu Y, Yin C, Jiang Y. Next-generation sequencing identified genetic variations in families with fetal non-syndromic atrioventricular septal defects. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3732-3743. [PMID: 31949757 PMCID: PMC6962893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 05/09/2018] [Indexed: 06/10/2023]
Abstract
Atrioventricular septal defects (AVSDs) account for approximately 5% of all congenital heart disease (CHD). About half of AVSDs are diagnosed in cases with trisomy 21 (Down's syndrome, DS). However, many AVSDs occur sporadically and manifest as non-syndromic. The pathogenesis is complex and has not yet been fully elucidated. In the present study, we applied two advanced applications of next-generation sequencing (NGS) to explore the genetic variations in families with fetal non-syndromic AVSDs. Our study was mainly divided into two steps: (1) low-pass whole-genome sequencing (WGS) was used to detect the genome-wide copy number variations (CNVs) for included subjects; (2) whole-exome sequencing (WES) was used to detect the gene mutations for the subjects without AVSD-associated CNVs. A total of 17 heterozygous de novo CNVs and 19 heterozygous de novo gene mutations were selected, and 15 candidate genes were involved in these variations. Among these heterozygous de novo variations, most have potential pathogenicity for AVSDs, but the others require further investigation to confirm their pathogenicity. Our study not only shows the genetic diversity and the etiological complexity of AVSDs but also shows the rationality and practicability of this sequential genetic detection and analysis strategy.
Collapse
Affiliation(s)
- Jinyu Xu
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Qingqing Wu
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Li Wang
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Jijing Han
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Yan Pei
- Department of Ultrasound, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Wenxue Zhi
- Department of Pathology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Yan Liu
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Chenghong Yin
- Department of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Yuxin Jiang
- Department of Ultrasound, Peking Union Medical College HospitalBeijing, China
| |
Collapse
|
24
|
Lygirou V, Latosinska A, Makridakis M, Mullen W, Delles C, Schanstra JP, Zoidakis J, Pieske B, Mischak H, Vlahou A. Plasma proteomic analysis reveals altered protein abundances in cardiovascular disease. J Transl Med 2018; 16:104. [PMID: 29665821 PMCID: PMC5905170 DOI: 10.1186/s12967-018-1476-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/06/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) describes the pathological conditions of the heart and blood vessels. Despite the large number of studies on CVD and its etiology, its key modulators remain largely unknown. To this end, we performed a comprehensive proteomic analysis of blood plasma, with the scope to identify disease-associated changes after placing them in the context of existing knowledge, and generate a well characterized dataset for further use in CVD multi-omics integrative analysis. METHODS LC-MS/MS was employed to analyze plasma from 32 subjects (19 cases of various CVD phenotypes and 13 controls) in two steps: discovery (13 cases and 8 controls) and test (6 cases and 5 controls) set analysis. Following label-free quantification, the detected proteins were correlated to existing plasma proteomics datasets (plasma proteome database; PPD) and functionally annotated (Cytoscape, Ingenuity Pathway Analysis). Differential expression was defined based on identification confidence (≥ 2 peptides per protein), statistical significance (Mann-Whitney p value ≤ 0.05) and a minimum of twofold change. RESULTS Peptides detected in at least 50% of samples per group were considered, resulting in a total of 3796 identified proteins (838 proteins based on ≥ 2 peptides). Pathway annotation confirmed the functional relevance of the findings (representation of complement cascade, fibrin clot formation, platelet degranulation, etc.). Correlation of the relative abundance of the proteins identified in the discovery set with their reported concentrations in the PPD was significant, confirming the validity of the quantification method. The discovery set analysis revealed 100 differentially expressed proteins between cases and controls, 39 of which were verified (≥ twofold change) in the test set. These included proteins already studied in the context of CVD (such as apolipoprotein B, alpha-2-macroglobulin), as well as novel findings (such as low density lipoprotein receptor related protein 2 [LRP2], protein SZT2) for which a mechanism of action is suggested. CONCLUSIONS This proteomic study provides a comprehensive dataset to be used for integrative and functional studies in the field. The observed protein changes reflect known CVD-related processes (e.g. lipid uptake, inflammation) but also novel hypotheses for further investigation including a potential pleiotropic role of LPR2 but also links of SZT2 to CVD.
Collapse
Affiliation(s)
- Vasiliki Lygirou
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, 115 27, Athens, Greece
| | | | - Manousos Makridakis
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, 115 27, Athens, Greece
| | - William Mullen
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institute of Cardiovascular and Metabolic Disease, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Jerome Zoidakis
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, 115 27, Athens, Greece
| | - Burkert Pieske
- Deutsches Herzzentrum Berlin, Augustenburger Pl. 1, 13353, Berlin, Germany
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, Rotenburger Straße 20, 30659, Hannover, Germany
| | - Antonia Vlahou
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephessiou Street, 115 27, Athens, Greece.
| |
Collapse
|
25
|
Zwier MV, Baardman ME, van Dijk TH, Jurdzinski A, Wisse LJ, Bloks VW, Berger RMF, DeRuiter MC, Groen AK, Plösch T. Maternal-fetal cholesterol transport in the second half of mouse pregnancy does not involve LDL receptor-related protein 2. Acta Physiol (Oxf) 2017; 220:471-485. [PMID: 28024118 DOI: 10.1111/apha.12845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/25/2016] [Accepted: 12/16/2016] [Indexed: 12/14/2022]
Abstract
AIM LDL receptor-related protein type 2 (LRP2) is highly expressed on both yolk sac and placenta. Mutations in the corresponding gene are associated with severe birth defects in humans, known as Donnai-Barrow syndrome. We here characterized the contribution of LRP2 and maternal plasma cholesterol availability to maternal-fetal cholesterol transport and fetal cholesterol levels in utero in mice. METHODS Lrp2+/- mice were mated heterozygously to yield fetuses of all three genotypes. Half of the dams received a 0.5% probucol-enriched diet during gestation to decrease maternal HDL cholesterol. At E13.5, the dams received an injection of D7-labelled cholesterol and were provided with 1-13 C acetate-supplemented drinking water. At E16.5, fetal tissues were collected and maternal cholesterol transport and fetal synthesis quantified by isotope enrichments in fetal tissues by GC-MS. RESULTS The Lrp2 genotype did not influence maternal-fetal cholesterol transport and fetal cholesterol. However, lowering of maternal plasma cholesterol levels by probucol significantly reduced maternal-fetal cholesterol transport. In the fetal liver, this was associated with increased cholesterol synthesis rates. No indications were found for an interaction between the Lrp2 genotype and maternal probucol treatment. CONCLUSION Maternal-fetal cholesterol transport and endogenous fetal cholesterol synthesis depend on maternal cholesterol concentrations but do not involve LRP2 in the second half of murine pregnancy. Our results suggest that the mouse fetus can compensate for decreased maternal cholesterol levels. It remains a relevant question how the delicate system of cholesterol transport and synthesis is regulated in the human fetus and placenta.
Collapse
Affiliation(s)
- M. V. Zwier
- Department of Pediatrics; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| | - M. E. Baardman
- Department of Genetics; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| | - T. H. van Dijk
- Department of Pediatrics; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
- Department of Laboratory Medicine; University of Groningen, University Medical Center Groningen; Groningen the Netherlands
| | - A. Jurdzinski
- Department of Pediatrics; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| | - L. J. Wisse
- Department of Anatomy and Embryology; Leiden University Medical Center; Leiden the Netherlands
| | - V. W. Bloks
- Department of Pediatrics; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| | - R. M. F. Berger
- Center for Congenital Heart Diseases; Beatrix Children's Hospital; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| | - M. C. DeRuiter
- Department of Anatomy and Embryology; Leiden University Medical Center; Leiden the Netherlands
| | - A. K. Groen
- Department of Pediatrics; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| | - T. Plösch
- Department of Obstetrics and Gynaecology; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| |
Collapse
|
26
|
Poelmann RE, Gittenberger-de Groot AC, Biermans MWM, Dolfing AI, Jagessar A, van Hattum S, Hoogenboom A, Wisse LJ, Vicente-Steijn R, de Bakker MAG, Vonk FJ, Hirasawa T, Kuratani S, Richardson MK. Outflow tract septation and the aortic arch system in reptiles: lessons for understanding the mammalian heart. EvoDevo 2017; 8:9. [PMID: 28491275 PMCID: PMC5424407 DOI: 10.1186/s13227-017-0072-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/03/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cardiac outflow tract patterning and cell contribution are studied using an evo-devo approach to reveal insight into the development of aorto-pulmonary septation. RESULTS We studied embryonic stages of reptile hearts (lizard, turtle and crocodile) and compared these to avian and mammalian development. Immunohistochemistry allowed us to indicate where the essential cell components in the outflow tract and aortic sac were deployed, more specifically endocardial, neural crest and second heart field cells. The neural crest-derived aorto-pulmonary septum separates the pulmonary trunk from both aortae in reptiles, presenting with a left visceral and a right systemic aorta arising from the unseptated ventricle. Second heart field-derived cells function as flow dividers between both aortae and between the two pulmonary arteries. In birds, the left visceral aorta disappears early in development, while the right systemic aorta persists. This leads to a fusion of the aorto-pulmonary septum and the aortic flow divider (second heart field population) forming an avian aorto-pulmonary septal complex. In mammals, there is also a second heart field-derived aortic flow divider, albeit at a more distal site, while the aorto-pulmonary septum separates the aortic trunk from the pulmonary trunk. As in birds there is fusion with second heart field-derived cells albeit from the pulmonary flow divider as the right 6th pharyngeal arch artery disappears, resulting in a mammalian aorto-pulmonary septal complex. In crocodiles, birds and mammals, the main septal and parietal endocardial cushions receive neural crest cells that are functional in fusion and myocardialization of the outflow tract septum. Longer-lasting septation in crocodiles demonstrates a heterochrony in development. In other reptiles with no indication of incursion of neural crest cells, there is either no myocardialized outflow tract septum (lizard) or it is vestigial (turtle). Crocodiles are unique in bearing a central shunt, the foramen of Panizza, between the roots of both aortae. Finally, the soft-shell turtle investigated here exhibits a spongy histology of the developing carotid arteries supposedly related to regulation of blood flow during pharyngeal excretion in this species. CONCLUSIONS This is the first time that is shown that an interplay of second heart field-derived flow dividers with a neural crest-derived cell population is a variable but common, denominator across all species studied for vascular patterning and outflow tract septation. The observed differences in normal development of reptiles may have impact on the understanding of development of human congenital outflow tract malformations.
Collapse
Affiliation(s)
- Robert E Poelmann
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands.,Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | | | - Marcel W M Biermans
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Anne I Dolfing
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Armand Jagessar
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Sam van Hattum
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Amanda Hoogenboom
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Lambertus J Wisse
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, Leiden, The Netherlands
| | - Rebecca Vicente-Steijn
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands.,Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, Leiden, The Netherlands
| | - Merijn A G de Bakker
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Freek J Vonk
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands.,Naturalis Biodiversity Center, Darwinweg 2, Leiden, The Netherlands
| | - Tatsuya Hirasawa
- Laboratory for Evolutionary Morphology, RIKEN, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Michael K Richardson
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| |
Collapse
|