1
|
Giordano E, Ponticelli I, Attard S, Pagano TB, Pisu MC. Ultrasound, Histomorphologic, and Immunohistochemical Analysis of a Cardiac Tumor with Increased Purkinje Cells Detected in a Canine Fetus 42 Days into Pregnancy. Vet Sci 2024; 11:216. [PMID: 38787187 PMCID: PMC11126094 DOI: 10.3390/vetsci11050216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
A seven-year-old healthy female Chow Chow was referred for pregnancy monitoring. Ultrasonography was used to evaluate all pregnancy and fetus parameters, and they were found to be normal. During the examination of the 42 day pregnant bitch, an unusual mass was seen in a fetus's heart. This fetus had a cardiac frequency of 273-300 beats, while the others had heart rates of 220-240 beats. Natural vaginal birth occurred at 63 days pregnant: the first two puppies were stillborn but perfectly formed, and the other three were alive and had optimal APGAR. In one of two deceased puppies, an unusual, reddish, smooth mass occupying the space in the heart was found through necroscopy. The organ was submitted for histological examination. Histopathology, immunohistochemical, and histochemical analyses all indicated a cardiac tumor with increased Purkinje cells. This type of tumor has been described in infants, swine, bearded seals, and deer but never in fetuses and neonates of dogs. To our knowledge, this is the first such case reported in veterinary medicine.
Collapse
Affiliation(s)
- Enrico Giordano
- Veterinary Clinic Giordano-Ponticelli, 80011 Acerra, Italy; (E.G.); (I.P.)
| | - Ignazio Ponticelli
- Veterinary Clinic Giordano-Ponticelli, 80011 Acerra, Italy; (E.G.); (I.P.)
| | - Simona Attard
- VRC—Veterinary Reference Center, 10138 Turin, Italy;
| | | | | |
Collapse
|
2
|
Grego-Bessa J, Gómez-Apiñaniz P, Prados B, Gómez MJ, MacGrogan D, de la Pompa JL. Nrg1 Regulates Cardiomyocyte Migration and Cell Cycle in Ventricular Development. Circ Res 2023; 133:927-943. [PMID: 37846569 PMCID: PMC10631509 DOI: 10.1161/circresaha.123.323321] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Cardiac ventricles provide the contractile force of the beating heart throughout life. How the primitive endocardium-layered myocardial projections called trabeculae form and mature into the adult ventricles is of great interest for biology and regenerative medicine. Trabeculation is dependent on the signaling protein Nrg1 (neuregulin-1). However, the mechanism of action of Nrg1 and its role in ventricular wall maturation are poorly understood. METHODS We investigated the functions and downstream mechanisms of Nrg1 signaling during ventricular chamber development using confocal imaging, transcriptomics, and biochemical approaches in mice with cardiac-specific inactivation or overexpression of Nrg1. RESULTS Analysis of cardiac-specific Nrg1 mutant mice showed that the transcriptional program underlying cardiomyocyte-oriented cell division and trabeculae formation depends on endocardial Nrg1 to myocardial ErbB2 (erb-b2 receptor tyrosine kinase 2) signaling and phospho-Erk (phosphorylated extracellular signal-regulated kinase; pErk) activation. Early endothelial loss of Nrg1 and reduced pErk activation diminished cardiomyocyte Pard3 and Crumbs2 (Crumbs Cell Polarity Complex Component 2) protein and altered cytoskeletal gene expression and organization. These alterations are associated with abnormal gene expression related to mitotic spindle organization and a shift in cardiomyocyte division orientation. Nrg1 is crucial for trabecular growth and ventricular wall thickening by regulating an epithelial-to-mesenchymal transition-like process in cardiomyocytes involving migration, adhesion, cytoskeletal actin turnover, and timely progression through the cell cycle G2/M phase. Ectopic cardiac Nrg1 overexpression and high pErk signaling caused S-phase arrest, sustained high epithelial-to-mesenchymal transition-like gene expression, and prolonged trabeculation, blocking compact myocardium maturation. Myocardial trabecular patterning alterations resulting from above- or below-normal Nrg1-dependent pErk activation were concomitant with sarcomere actin cytoskeleton disorganization. The Nrg1 loss- and gain-of-function transcriptomes were enriched for Yap1 (yes-associated protein-1) gene signatures, identifying Yap1 as a potential downstream effector. Furthermore, biochemical and imaging data reveal that Nrg1 influences pErk activation and Yap1 nuclear-cytoplasmic distribution during trabeculation. CONCLUSIONS These data establish the Nrg1-ErbB2/ErbB4-Erk axis as a crucial regulator of cardiomyocyte cell cycle progression and migration during ventricular development.
Collapse
Affiliation(s)
- Joaquim Grego-Bessa
- Intercellular Signalling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (J.G.-B., P.G.-A., B.P., D.M., J.L.d.l.P.)
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain (J.G.-B., P.G.-A., B.P., D.M., J.L.d.l.P.)
| | - Paula Gómez-Apiñaniz
- Intercellular Signalling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (J.G.-B., P.G.-A., B.P., D.M., J.L.d.l.P.)
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain (J.G.-B., P.G.-A., B.P., D.M., J.L.d.l.P.)
| | - Belén Prados
- Intercellular Signalling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (J.G.-B., P.G.-A., B.P., D.M., J.L.d.l.P.)
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain (J.G.-B., P.G.-A., B.P., D.M., J.L.d.l.P.)
| | | | - Donal MacGrogan
- Intercellular Signalling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (J.G.-B., P.G.-A., B.P., D.M., J.L.d.l.P.)
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain (J.G.-B., P.G.-A., B.P., D.M., J.L.d.l.P.)
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (J.G.-B., P.G.-A., B.P., D.M., J.L.d.l.P.)
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain (J.G.-B., P.G.-A., B.P., D.M., J.L.d.l.P.)
| |
Collapse
|
3
|
Ivanova VV, Milto IV, Serebrjakova ON, Sukhodolo IV. The Rat Heart in the Prenatal and Postnatal Periods of Ontogenesis. Russ J Dev Biol 2021. [DOI: 10.1134/s1062360421050039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
4
|
Colpan M, Iwanski J, Gregorio CC. CAP2 is a regulator of actin pointed end dynamics and myofibrillogenesis in cardiac muscle. Commun Biol 2021; 4:365. [PMID: 33742108 PMCID: PMC7979805 DOI: 10.1038/s42003-021-01893-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 02/22/2021] [Indexed: 01/31/2023] Open
Abstract
The precise assembly of actin-based thin filaments is crucial for muscle contraction. Dysregulation of actin dynamics at thin filament pointed ends results in skeletal and cardiac myopathies. Here, we discovered adenylyl cyclase-associated protein 2 (CAP2) as a unique component of thin filament pointed ends in cardiac muscle. CAP2 has critical functions in cardiomyocytes as it depolymerizes and inhibits actin incorporation into thin filaments. Strikingly distinct from other pointed-end proteins, CAP2's function is not enhanced but inhibited by tropomyosin and it does not directly control thin filament lengths. Furthermore, CAP2 plays an essential role in cardiomyocyte maturation by modulating pre-sarcomeric actin assembly and regulating α-actin composition in mature thin filaments. Identification of CAP2's multifunctional roles provides missing links in our understanding of how thin filament architecture is regulated in striated muscle and it reveals there are additional factors, beyond Tmod1 and Lmod2, that modulate actin dynamics at thin filament pointed ends.
Collapse
Affiliation(s)
- Mert Colpan
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Jessika Iwanski
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
5
|
Liu HX, Jing YX, Wang JJ, Yang YP, Wang YX, Li HR, Song L, Li AH, Cui HL, Jing Y. Expression patterns of intermediate filament proteins desmin and lamin A in the developing conduction system of early human embryonic hearts. J Anat 2019; 236:540-548. [PMID: 31670395 DOI: 10.1111/joa.13108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2019] [Indexed: 11/27/2022] Open
Abstract
Since embryonic heart development is a complex process and acquisition of human embryonic specimens is challenging, the mechanism by which the embryonic conduction system develops remains unclear. Herein, we attempt to gain insights into this developmental process through immunohistochemical staining and 3D reconstructions. Expression analysis of T-box transcription factor 3, cytoskeleton desmin, and nucleoskeleton lamin A protein in human embryos in Carnegie stages 11-20 showed that desmin is preferentially expressed in the myocardium of the central conduction system compared with the peripheral conduction system, and is co-expressed with T-box transcription factor 3 in the central conduction system. Further, lamin A was first expressed in the embryonic ventricular trabeculations, where the terminal ramifications of the peripheral conduction system develop, and extended progressively to all parts of the central conduction system. The uncoupled spatiotemporal distribution pattern of lamin A and desmin indicated that the association of cytoskeleton desmin and nucleoskeleton lamin A may be a late event in human embryonic heart development. Compared with model animals, our data provide a direct morphological basis for understanding the arrhythmogenesis caused by mutations in human DES and LMNA genes.
Collapse
Affiliation(s)
- Hui-Xia Liu
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yi-Xin Jing
- Department of Internal Medicine, Shenzhen Nanshan People's Hospital, Shenzhen, Guangdong, China
| | - Jing-Jing Wang
- 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
| | - Yun-Xiu Wang
- Department of Obstetrics and Gynaecology, Children's Hospital of Shanxi, Taiyuan, Shanxi, China
| | - Hai-Rong Li
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Song
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ai-Hong Li
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hui-Lin Cui
- 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
|
6
|
The human phrenic nerve serves as a morphological conduit for autonomic nerves and innervates the caval body of the diaphragm. Sci Rep 2018; 8:11697. [PMID: 30076368 PMCID: PMC6076324 DOI: 10.1038/s41598-018-30145-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/16/2018] [Indexed: 01/08/2023] Open
Abstract
Communicating fibres between the phrenic nerve and sympathetic nervous system may exist, but have not been characterized histologically and immunohistochemically, even though increased sympathetic activity due to phrenic nerve stimulation for central sleep apnoea may entail morbidity and mortality. We, therefore, conducted a histological study of the phrenic nerve to establish the presence of catecholaminergic fibres throughout their course. The entire phrenic nerves of 35 formalin-fixed human cadavers were analysed morphometrically and immunohistochemically. Furthermore, the right abdominal phrenic nerve was serially sectioned and reconstructed. The phrenic nerve contained 3 ± 2 fascicles in the neck that merged to form a single fascicle in the thorax and split again into 3 ± 3 fascicles above the diaphragm. All phrenic nerves contained catecholaminergic fibres, which were distributed homogenously or present as distinct areas within a fascicle or as separate fascicles. The phrenicoabdominal branch of the right phrenic nerve is a branch of the celiac plexus and, therefore, better termed the “phrenic branch of the celiac plexus”. The wall of the inferior caval vein in the diaphragm contained longitudinal strands of myocardium and atrial natriuretic peptide-positive paraganglia (“caval bodies”) that where innervated by the right phrenic nerve.
Collapse
|
7
|
Lenčo J, Lenčová-Popelová O, Link M, Jirkovská A, Tambor V, Potůčková E, Stulík J, Šimůnek T, Štěrba M. Proteomic investigation of embryonic rat heart-derived H9c2 cell line sheds new light on the molecular phenotype of the popular cell model. Exp Cell Res 2015; 339:174-86. [DOI: 10.1016/j.yexcr.2015.10.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 10/20/2015] [Accepted: 10/23/2015] [Indexed: 01/15/2023]
|
8
|
Sakurai M, Kuninaga N, Takeuchi T, Tsuka T, Morita T. Congenital multifocal increase of Purkinje fibres in a calf with cardiac conduction delay. J Comp Pathol 2014; 151:234-7. [PMID: 25084712 DOI: 10.1016/j.jcpa.2014.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 05/29/2014] [Accepted: 06/05/2014] [Indexed: 11/17/2022]
Abstract
A female 4-month-old Holstein-Friesian calf was presented in heart failure. Microscopical examination of samples of the cardiac wall taken at necropsy examination revealed numerous aggregates of Purkinje fibres, particularly in the perivascular areas. Some Purkinje fibres were stained strongly with phosphotungstic acid haematoxylin and immunohistochemically were shown to express alpha smooth muscle actin, indicating an embryonic-like Purkinje fibre phenotype. A diagnosis of congenital multifocal increase of Purkinje fibres was made. The histological features of this case resemble multifocal cardiac Purkinje cell tumour of the heart in man.
Collapse
Affiliation(s)
- M Sakurai
- Department of Veterinary Pathology, Tottori University, Tottori, Japan
| | - N Kuninaga
- Department of Veterinary Pathology, Tottori University, Tottori, Japan
| | - T Takeuchi
- Department of Veterinary Laboratory Medicine, Tottori University, Tottori, Japan
| | - T Tsuka
- Department of Veterinary Diagnostic Imaging, Tottori University, Tottori, Japan
| | - T Morita
- Department of Veterinary Pathology, Tottori University, Tottori, Japan.
| |
Collapse
|
9
|
Varadkar P, Despres D, Kraman M, Lozier J, Phadke A, Nagaraju K, Mccright B. The protein phosphatase 2A B56γ regulatory subunit is required for heart development. Dev Dyn 2014; 243:778-90. [PMID: 24425002 DOI: 10.1002/dvdy.24111] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 12/16/2013] [Accepted: 12/19/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Protein Phosphatase 2A (PP2A) function is controlled by regulatory subunits that modulate the activity of the catalytic subunit and direct the PP2A complex to specific intracellular locations. To study PP2A's role in signal transduction pathways that control growth and differentiation in vivo, a transgenic mouse lacking the B56γ regulatory subunit of PP2A was made. RESULTS Lack of PP2A activity specific to the PP2A-B56γ holoenzyme, resulted in the formation of an incomplete ventricular septum and a decrease in the number of ventricular cardiomyocytes. During cardiac development, B56γ is expressed in the nucleus of α-actinin-positive cardiomyocytes that contain Z-bands. The pattern of B56γ expression correlated with the cardiomyocyte apoptosis we observed in B56γ-deficient mice during mid to late gestation. In addition to the cardiac phenotypes, mice lacking B56γ have a decrease in locomotive coordination and gripping strength, indicating that B56γ has a role in controlling PP2A activity required for efficient neuromuscular function. CONCLUSIONS PP2A-B56γ activity is required for efficient cardiomyocyte maturation and survival. The PP2A B56γ regulatory subunit controls PP2A substrate specificity in vivo in a manner that cannot be fully compensated for by other B56 subunits.
Collapse
Affiliation(s)
- Prajakta Varadkar
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, FDA, Bethesda, Maryland
| | | | | | | | | | | | | |
Collapse
|
10
|
Berry SE, Andruszkiewicz P, Chun JL, Hong J. Nestin expression in end-stage disease in dystrophin-deficient heart: implications for regeneration from endogenous cardiac stem cells. Stem Cells Transl Med 2013; 2:848-61. [PMID: 24068741 PMCID: PMC3808200 DOI: 10.5966/sctm.2012-0174] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 05/28/2013] [Indexed: 01/16/2023] Open
Abstract
Nestin(+) cardiac stem cells differentiate into striated cells following myocardial infarct. Transplantation of exogenous stem cells into myocardium of a murine model for Duchenne muscular dystrophy (DMD) increased proliferation of endogenous nestin(+) stem cells and resulted in the appearance of nestin(+) striated cells. This correlated with, and may be responsible for, prevention of dilated cardiomyopathy. We examined nestin(+) stem cells in the myocardium of dystrophin/utrophin-deficient (mdx/utrn(-/-)) mice, a model for DMD. We found that 92% of nestin(+) interstitial cells expressed Flk-1, a marker present on cardiac progenitor cells that differentiate into the cardiac lineage, and that a subset expressed Sca-1, present on adult cardiac cells that become cardiomyocytes. Nestin(+) interstitial cells maintained expression of Flk-1 but lost Sca-1 expression with age and were present in lower numbers in dystrophin-deficient heart than in wild-type heart. Unexpectedly, large clusters of nestin(+) striated cells ranging in size from 20 to 250 cells and extending up to 500 μm were present in mdx/utrn(-/-) heart near the end stage of disease. These cells were also present in dystrophin-deficient mdx/utrn(+/-) and mdx heart but not wild-type heart. Nestin(+) striated cells expressed cardiac troponin I, desmin, and Connexin 43 and correlated with proinflammatory CD68(+) macrophages. Elongated nestin(+) interstitial cells with striations were observed that did not express Flk-1 or the late cardiac marker cardiac troponin I but strongly expressed the early cardiac marker desmin. Nestin was also detected in endothelial and smooth muscle cells. These data indicate that new cardiomyocytes form in dystrophic heart, and nestin(+) interstitial cells may generate them in addition to other cells of the cardiac lineage.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Ly/genetics
- Antigens, Ly/metabolism
- Biomarkers/metabolism
- Connexin 43/genetics
- Connexin 43/metabolism
- Disease Models, Animal
- Dystrophin/deficiency
- Dystrophin/genetics
- Dystrophin/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/physiology
- Heart/physiopathology
- Macrophages/metabolism
- Macrophages/physiology
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
- Myocardial Infarction/genetics
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/physiology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Nestin/biosynthesis
- Nestin/genetics
- Nestin/metabolism
- Regeneration/genetics
- Regeneration/physiology
- Stem Cells/metabolism
- Stem Cells/physiology
- Utrophin/deficiency
- Utrophin/genetics
- Utrophin/metabolism
- Vascular Endothelial Growth Factor Receptor-2/genetics
- Vascular Endothelial Growth Factor Receptor-2/metabolism
Collapse
Affiliation(s)
- Suzanne E. Berry
- Department of Comparative Biosciences
- Institute for Genomic Biology
- Neuroscience Program, and
| | | | - Ju Lan Chun
- Department of Animal Sciences, University of Illinois, Urbana, Illinois, USA
| | - Jun Hong
- Department of Comparative Biosciences
| |
Collapse
|
11
|
Rana MS, Sizarov A, Christoffels VM, Moorman AFM. Development of the human aortic arch system captured in an interactive three-dimensional reference model. Am J Med Genet A 2013; 164A:1372-83. [PMID: 23613216 DOI: 10.1002/ajmg.a.35881] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 12/31/2012] [Indexed: 11/12/2022]
Abstract
Variations and mutations in the human genome, such as 22q11.2 microdeletion, can increase the risk for congenital defects, including aortic arch malformations. Animal models are increasingly expanding our molecular and genetic insights into aortic arch development. However, in order to justify animal-to-human extrapolations, a human morphological, and molecular reference model would be of great value, but is currently lacking. Here, we present interactive three-dimensional reconstructions of the developing human aortic arch system, supplemented with the protein distribution of developmental markers for patterning and growth, including T-box transcription factor TBX1, a major candidate for the phenotypes found in patients with the 22q11.2 microdeletion. These reconstructions and expression data facilitate unbiased interpretations, and reveal previously unappreciated aspects of human aortic arch development. Based on our reconstructions and on reported congenital anomalies of the pulmonary trunk and tributaries, we postulate that the pulmonary arteries originate from the aortic sac, rather than from the sixth pharyngeal arch arteries. Similar to mouse, TBX1 is expressed in pharyngeal mesenchyme and epithelia. The endothelium of the pharyngeal arch arteries is largely negative for TBX1 and family member TBX2 but expresses neural crest marker AP2α, which gradually decreases with ongoing development of vascular smooth muscle. At early stages, the pharyngeal arch arteries, aortic sac, and the dorsal aortae in particular were largely negative for proliferation marker Ki67, potentially an important parameter during aortic arch system remodeling. Together, our data support current animal-to-human extrapolations and future genetic and molecular analyses using animal models of congenital heart disease. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- M Sameer Rana
- Heart Failure Research Center, Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
12
|
Saravanakumar M, Devaraj H. Notch signalling in cardiovasculogenesis: insight into their role in early cardiovascular development. Mol Biol Rep 2012; 40:3537-47. [DOI: 10.1007/s11033-012-2427-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 12/18/2012] [Indexed: 11/29/2022]
|
13
|
Fujimoto KL, Tobita K, Guan J, Hashizume R, Takanari K, Alfieri CM, Yutzey KE, Wagner WR. Placement of an elastic biodegradable cardiac patch on a subacute infarcted heart leads to cellularization with early developmental cardiomyocyte characteristics. J Card Fail 2012; 18:585-95. [PMID: 22748493 DOI: 10.1016/j.cardfail.2012.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 05/16/2012] [Accepted: 05/18/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Placement of an elastic biodegradable patch onto a subacute myocardial infarct (MI) provides temporary elastic support that may act to effectively alter adverse left ventricular (LV) remodeling processes. METHODS Two weeks after permanent left coronary ligation in Lewis rats, the infarcted anterior wall was covered with polyester urethane urea (MI + PEUU; n = 15) or expanded polytetrafluoroethylene (MI + ePTFE; n = 15) patches, or had no implantation (MI + sham; n = 12). Eight weeks after surgery, cardiac function and histology were assessed. RESULTS The ventricular wall in the MI + ePTFE and MI + sham groups was composed of fibrous tissue, whereas PEUU implantation induced α-smooth muscle actin-positive muscle bundles coexpressing sarcomeric α-actinin and cardiac-specific troponin-T. This pattern of colocalization was also found in developing embryonic myocardium. Cardiac transcription factors Nkx-2.5 and GATA-4 were strongly expressed in the muscle bundles. In the MI + sham group, end-diastolic LV cavity area (EDA) increased and the percentage of fractional area change (%FAC) decreased. For ePTFE patched animals, both EDA and %FAC decreased. In contrast, with MI + PEUU patching, %FAC increased and EDA was maintained. With dobutamine-stress echocardiography, MI + PEUU patched LVs possessed contractile reserve significantly larger than the MI + sham group. CONCLUSIONS MI + PEUU patch implantation onto subacute infarcted myocardium induced muscle cellularization with characteristics of early developmental cardiomyocytes as well as providing a functional reserve.
Collapse
Affiliation(s)
- Kazuro L Fujimoto
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Imamura T, Ishizuka O, Lei Z, Hida S, Sudha GS, Kato H, Nishizawa O. Bone Marrow-Derived Cells Implanted into Radiation-Injured Urinary Bladders Reconstruct Functional Bladder Tissues in Rats. Tissue Eng Part A 2012; 18:1698-709. [DOI: 10.1089/ten.tea.2012.0061] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Tetsuya Imamura
- Department of Lower Urinary Tract Medicine, Shinshu University School of Medicine, Nagano, Japan
| | - Osamu Ishizuka
- Department of Lower Urinary Tract Medicine, Shinshu University School of Medicine, Nagano, Japan
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan
| | - Zhang Lei
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan
| | - Shigeaki Hida
- Department of Molecular Oncology, Institute on Aging and Adaptation, Shinshu University Graduate School of Medicine, Nagano, Japan
| | | | - Haruaki Kato
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan
| | - Osamu Nishizawa
- Department of Lower Urinary Tract Medicine, Shinshu University School of Medicine, Nagano, Japan
- Department of Urology, Shinshu University School of Medicine, Nagano, Japan
| |
Collapse
|
15
|
Abstract
Muscularis externa of mouse esophagus is composed of two skeletal muscle layers in the adult. But less attention is paid to the histogenesis of the muscularis externa of the esophagus, and controversies still exist about the developmental process and the spatio-temporal expression characteristics of muscle-specific proteins during the development of esophageal muscularis externa. To further probe into the developmental pattern of muscularis externa of the mouse esophagus and the expression characteristics of different muscle-specific proteins, immunohistochemical and terminal deoxyribonucleotidyl transferase-mediated deoxyuridine triphosphate (dUTP)-digoxigenin nick-end labeling apoptotic staining methods are used to investigate the expression patterns of different muscle-specific proteins and to elucidate the relationship of these protein expressions with the development of muscularis externa of the mouse esophagus. Thus, an understanding of the developing esophageal muscularis externa may be important for developing therapeutic strategies for the treatment of human esophagus diseases. Serial sections of mouse embryos from embryonic day (ED) 12 to ED18, and full-length esophagi from postnatal first to 5th day were stained with monoclonal antibodies against α-smooth muscle actin (α-SMA), α-sarcomerical actin (α-SCA), desmin, and monoclonal anti-skeletal myosin (MHC), while apoptosis was determined using the terminal deoxyribonucleotidyl transferase-mediated dUTP-digoxigenin nick-end labeling assay. The expression of α-SMA was started at ED12. During the development of ED14-ED15, α-SMA positive cells were seen extending from the walls of left three, four, and six arch arteries toward the dorsal wall of esophagus. Stronger expression of α-SCA and desmin could be detected at ED14 and ED15, expression intensity in caudal segment and inner layer was stained stronger than that of cranial segment and outer layer, but after ED16, strong expression of α-SCA and desmin was found in the outer layer of muscularis externa. Expression of MHC was first detected in the outer layer of cranial segment of muscularis externa at ED17. At ED18, MHC had extended to the level of thyroid gland, staining intensity in the outer layer and cranial segment was stronger than that of inner layer and caudal segment. One to five days after birth, the thickness of the esophageal muscle layer was obviously increased. Most of the muscle cells in the cranial segment of esophagus showed strong expression of α-SCA and clear cross striations at higher magnification. With progression toward the caudal segment, expression intensity of α-SCA became weaker, but the expression intensity of desmin was the same at different levels of esophagus. The muscle fibers were arranged densely with high expression of MHC in the cranial segment. During the development of esophageal muscularis externa, few apoptotic cells were observed. α-SMA, α-SCA, desmin, and MHC show different expression patterns. The differentiation of outer layer of esophageal muscularis externa is quicker than that of inner layer, and the caudal segment is quicker than that of the cranial segment. Besides, apoptosis may not participate in the development of esophageal muscularis externa. The smooth muscle cells from arch arteries may participate in the development of esophageal muscularis externa.
Collapse
Affiliation(s)
- X-M Cao
- Department of Histology and Embryology, Shanxi Medical University, Taiyuan, China
| | | | | | | | | |
Collapse
|
16
|
Rémond MC, Iaffaldano G, O'Quinn MP, Mezentseva NV, Garcia V, Harris BS, Gourdie RG, Eisenberg CA, Eisenberg LM. GATA6 reporter gene reveals myocardial phenotypic heterogeneity that is related to variations in gap junction coupling. Am J Physiol Heart Circ Physiol 2011; 301:H1952-64. [PMID: 21908788 DOI: 10.1152/ajpheart.00635.2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This study examined transgenic mice whose expression of a β-galactosidase (lacZ) reporter is driven by a GATA6 gene enhancer. Previous investigations established that transcription of the transgene was associated with precardiac mesoderm and primary heart tube myocardium, which decreased progressively, so that its expression was no longer observed within ventricular myocardium by midgestation. Expression of this reporter in the adult was investigated for insights into myocyte homeostasis and cardiovascular biology. Morphometric analysis determined that <1% of myocytes, often found in small clusters, express this GATA6-associated reporter in the adult heart. LacZ expression was also found in the ascending aorta. Myocardial expression of the transgene was not associated with a proliferative phenotype or new myocyte formation, as lacZ-positive myocytes neither labeled with cell division markers nor following 5-bromodeoxyuridine pulse-chase experimentation. Despite exhibiting normal adherens junctions, these myocytes appeared to exhibit decreased connexin 43 gap junctions. Treatment with the gap junctional blocker heptanol both in vivo and in culture elevated myocardial β-galactosidase activity, suggesting that deficient gap junctional communication underlies expression of the transgenic reporter. LacZ expression within the myocardium was also enhanced in response to cryoinjury and isoproterenol-induced hypertrophy. These results reveal a previously uncharacterized phenotypic heterogeneity in the myocardium and suggest that decreased gap junctional coupling leads to induction of a signaling pathway that utilizes a unique GATA6 enhancer. Upregulation of lacZ reporter gene expression following cardiac injury indicates this transgenic mouse may serve as a model for examining the transition of the heart from healthy to pathological states.
Collapse
Affiliation(s)
- Mathieu C Rémond
- New York Medical College/Westchester Medical Center Stem Cell Laboratory, Department of Physiology, New York Medical College, Valhalla, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Yamamoto M, Abe SI, Rodríguez-Vázquez JF, Fujimiya M, Murakami G, Ide Y. Immunohistochemical distribution of desmin in the human fetal heart. J Anat 2011; 219:253-8. [PMID: 21496015 PMCID: PMC3162244 DOI: 10.1111/j.1469-7580.2011.01382.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2011] [Indexed: 11/29/2022] Open
Abstract
Desmin is a member of the intermediate filaments, which play crucial roles in the maturation, maintenance and recovery of muscle fibers. Its expression has been examined in human cardiac muscle, rat and chicken, but its spatial distribution in the human fetal heart has not been described. The present study investigated desmin expression in the human fetal heart and associated great vessels in 14 mid-term fetuses from 9 to 18 weeks of gestation. Immunoreactivity for myosin heavy chain (MHC) and alpha smooth muscle actin (α-SMA), as well as neuron-specific enolase (NSE), was also examined. Increased expression of desmin from 9 to 18 weeks was clearly localized in the atrial wall, the proximal portions of the pulmonary vein and vena cava, and around the atrioventricular node. Desmin-positive structures were also positive for MHC. Meanwhile, the great vessels were also positive for α-SMA. The distribution of desmin exhibited a pattern quite different from that described in previous studies of rat and chicken. Thus, desmin in the human fetal heart does not seem to play a general role in myocardial differentiation but rather a specific role closely related to the maturation of the α-isozyme of MHC. Desmin expression in the developing fetal heart also appeared to be induced by mechanical stress due to the involvement of venous walls against the atrium.
Collapse
|
18
|
Potta SP, Sheng X, Gaspar JA, Meganathan K, Jagtap S, Pfannkuche K, Winkler J, Hescheler J, Papadopoulos S, Sachinidis A. Functional Characterization and Gene Expression Profiling of α-Smooth Muscle Actin Expressing Cardiomyocytes Derived from Murine Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2011; 8:229-42. [DOI: 10.1007/s12015-011-9271-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
19
|
Grimes AC, Durán AC, Sans-Coma V, Hami D, Santoro MM, Torres M. Phylogeny informs ontogeny: a proposed common theme in the arterial pole of the vertebrate heart. Evol Dev 2011; 12:552-67. [PMID: 21040422 DOI: 10.1111/j.1525-142x.2010.00441.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In chick and mouse embryogenesis, a population of cells described as the secondary heart field (SHF) adds both myocardium and smooth muscle to the developing cardiac outflow tract (OFT). Following this addition, at approximately HH stage 22 in chick embryos, for example, the SHF can be identified architecturally by an overlapping seam at the arterial pole, where beating myocardium forms a junction with the smooth muscle of the arterial system. Previously, using either immunohistochemistry or nitric oxide indicators such as diaminofluorescein 2-diacetate, we have shown that a similar overlapping architecture also exists in the arterial pole of zebrafish and some shark species. However, although recent work suggests that development of the zebrafish OFT may also proceed by addition of a SHF-like population of cells, the presence of a true SHF in zebrafish and in many other developmental biological models remains an open question. We performed a comprehensive morphological study of the OFT of a wide range of vertebrates. Our data suggest that all vertebrates possess three fundamental OFT components: a proximal myocardial component, a distal smooth muscle component, and a middle component that contains overlapping myocardium and smooth muscle surrounding and supporting the outflow valves. Because the middle OFT component of avians and mammals is derived from the SHF, our observations suggest that a SHF may be an evolutionarily conserved theme in vertebrate embryogenesis.
Collapse
Affiliation(s)
- Adrian C Grimes
- Departamento de Biología del Desarrollo Cardiovascular, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
20
|
Ambler CA, Watt FM. Adult epidermal Notch activity induces dermal accumulation of T cells and neural crest derivatives through upregulation of jagged 1. Development 2010; 137:3569-79. [PMID: 20940224 PMCID: PMC2964092 DOI: 10.1242/dev.050310] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2010] [Indexed: 12/13/2022]
Abstract
Notch signalling regulates epidermal differentiation and tumour formation via non-cell autonomous mechanisms that are incompletely understood. This study shows that epidermal Notch activation via a 4-hydroxy-tamoxifen-inducible transgene caused epidermal thickening, focal detachment from the underlying dermis and hair clumping. In addition, there was dermal accumulation of T lymphocytes and stromal cells, some of which localised to the blisters at the epidermal-dermal boundary. The T cell infiltrate was responsible for hair clumping but not for other Notch phenotypes. Notch-induced stromal cells were heterogeneous, expressing markers of neural crest, melanocytes, smooth muscle and peripheral nerve. Although Slug1 expression was expanded in the epidermis, the stromal cells did not arise through epithelial-mesenchymal transition. Epidermal Notch activation resulted in upregulation of jagged 1 in both epidermis and dermis. When Notch was activated in the absence of epidermal jagged 1, jagged 1 was not upregulated in the dermis, and epidermal thickening, blister formation, accumulation of T cells and stromal cells were inhibited. Gene expression profiling revealed that epidermal Notch activation resulted in upregulation of several growth factors and cytokines, including TNFα, the expression of which was dependent on epidermal jagged 1. We conclude that jagged 1 is a key mediator of non-cell autonomous Notch signalling in skin.
Collapse
Affiliation(s)
- Carrie A. Ambler
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Cambridge CB2 0RE, UK
- School of Biological and Biomedical Sciences and NorthEast England Stem Cell Institute, Durham University, South Road, Durham DH1 3LE, UK
| | - Fiona M. Watt
- Cancer Research UK, Cambridge Research Institute, Li Ka Shing Centre, Cambridge CB2 0RE, UK
- Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
21
|
The area composita of adhering junctions connecting heart muscle cells of vertebrates. VII. The different types of lateral junctions between the special cardiomyocytes of the conduction system of ovine and bovine hearts. Eur J Cell Biol 2010; 89:365-78. [DOI: 10.1016/j.ejcb.2009.11.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 10/29/2009] [Accepted: 11/09/2009] [Indexed: 11/19/2022] Open
|
22
|
IMAMURA T, ISHIZUKA O, YAMAMOTO T, GOTOH M, NISHIZAWA O. Bone Marrow-Derived Cells Implanted into Freeze-Injured Urinary Bladders Reconstruct Functional Smooth Muscle Layers. Low Urin Tract Symptoms 2010; 2:1-10. [DOI: 10.1111/j.1757-5672.2010.00066.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
23
|
Parekh A, Long RA, Iannone EC, Chancellor MB, Sacks MS. Assessing the effects of transforming growth factor-beta1 on bladder smooth muscle cell phenotype. I. Modulation of in vitro contractility. J Urol 2009; 182:1210-5. [PMID: 19625042 DOI: 10.1016/j.juro.2009.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Indexed: 10/20/2022]
Abstract
PURPOSE Modulation of the bladder smooth muscle cell phenotype contributes to the resulting bladder dysfunction in many pathological bladder conditions. Transforming growth factor-beta1 is an important regulator of cellular phenotype in fibrotic diseases that has specific effects on bladder smooth muscle cells associated with phenotypic changes. We verified transforming growth factor-beta1 expression in neurogenic bladder tissue and investigated its effects on bladder smooth muscle cell collagen gel contraction. MATERIALS AND METHODS Transforming growth factor-beta1 immunostaining was performed on tissue sections from spinalized rats and quantified based on the ratio of fluorescence to total detrusor area. Rat bladder smooth muscle cells were seeded at different densities on anchored collagen gels and the effect of transforming growth factor-beta1 on contractility was assessed by measuring changes in the collagen gel area with time. Phenotypic changes induced by transforming growth factor-beta1 were detected by immunostaining for caldesmon and the specific isoform high molecular weight caldesmon. RESULTS Transforming growth factor-beta1 immunostaining revealed increased levels specifically in the detrusor of spinal cord injured rats. Rat bladder smooth muscle cell contraction increased with larger cell populations and was inhibited by transforming growth factor-beta1. Transforming growth factor-beta1 induced a decrease in high molecular weight caldesmon expression in bladder smooth muscle cells. CONCLUSIONS Increased transforming growth factor-beta1 expression in the detrusor of spinal cord injured rats implies up-regulation and localized signaling in response to injury. Bladder smooth muscle cells showed a loss of contractility in response to transforming growth factor-beta1 in all cell populations. A shift in phenotype was confirmed by high molecular weight caldesmon immunostaining. These results suggest that transforming growth factor-beta1 can modulate bladder smooth muscle cell function and may be a crucial regulator of bladder smooth muscle cell phenotype in pathological bladder conditions.
Collapse
Affiliation(s)
- Aron Parekh
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | | | | | | | | |
Collapse
|
24
|
Generality of a power-law long-term correlation in beat timings of single cardiac cells. Biochem Biophys Res Commun 2009; 387:19-24. [PMID: 19501567 DOI: 10.1016/j.bbrc.2009.05.143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 05/31/2009] [Indexed: 11/23/2022]
Abstract
Statistical properties of spontaneous contractions of atrial muscle cells were examined and compared to those of ventricular muscle cells. The cells derived from atria of neonatal rats exhibit spindle morphology, and they were found to express alpha-smooth muscle actin and hyperpolarization-activated cation channel 4, both of which are known marker of neonatal atrial muscle cells. The short-term properties of spontaneous contractions of atrial cells, characterized by considerably large beat rate and absence of bursts, are distinct from those of ventricular muscle cells. Despite of these differences, the long-term properties of the beat-rate fluctuations exhibit a remarkable similarity to those of ventricular cells. In particular, the presence of power-law correlation characterized as 1/f(beta) noise (beta approximately 1) was also confirmed for atrial cells for the first time. The observed similarity of the long-term characteristics of beat-rate fluctuation suggests the presence of a general regulatory mechanism of the cellular function.
Collapse
|
25
|
Orlandi A, Hao H, Ferlosio A, Clément S, Hirota S, Spagnoli LG, Gabbiani G, Chaponnier C. Alpha actin isoforms expression in human and rat adult cardiac conduction system. Differentiation 2009; 77:360-8. [PMID: 19281784 DOI: 10.1016/j.diff.2008.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 12/16/2008] [Accepted: 12/18/2008] [Indexed: 11/26/2022]
Abstract
In the adult heart, cardiac muscle comprises the working myocardium and the conduction system (CS). The latter includes the sinoatrial node (SAN), the internodal tract or bundle (IB), the atrioventricular node (AVN), the atrioventricular bundle (AVB), the bundle branches (BB) and the peripheral Purkinje fibers (PF). Most of the information concerning the phenotypic features of CS tissue derives from the characterization of avian and rodent developing hearts; data concerning the expression of actin isoforms in adult CS cardiomyocytes are scarce. Using specific antibodies, we investigated the distribution of alpha-skeletal (alpha-SKA), alpha-cardiac (alpha-CA), alpha-smooth muscle (alpha-SMA) actin isoforms and other muscle-typical proteins in the CS of human and rat hearts at different ages. SAN and IB cardiomyocytes were characterized by the presence of alpha-SMA, alpha-CA, calponin and caldesmon, whereas alpha-SKA and vimentin were absent. Double immunofluorescence demonstrated the co-localisation of alpha-SMA and alpha-CA in I-bands of SAN cardiomyocytes. AVN, AVB, BB and PF cardiomyocytes were alpha-SMA, calponin, caldesmon and vimentin negative, and alpha-CA and alpha-SKA positive. No substantial differences in actin isoform distribution were observed in human and rat hearts, except for the presence of isolated subendocardial alpha-SMA positive cardiomyocytes co-expressing alpha-CA in the ventricular septum of the rat. Aging did not influence CS cardiomyocyte actin isoform expression profile. These findings support the concept that cardiomyocytes of SAN retain the phenotype of a developing myogenic cell throughout the entire life span.
Collapse
Affiliation(s)
- Augusto Orlandi
- Institute of Anatomic Pathology, Department of Biopathology and Image Diagnostics, Tor Vergata University of Rome-PTV, Via Montpellier 1, 00133, Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Imamura T, Kinebuchi Y, Ishizuka O, Seki S, Igawa Y, Nishizawa O. Implanted Mouse Bone Marrow-Derived Cells Reconstruct Layered Smooth Muscle Structures in Injured Urinary Bladders. Cell Transplant 2008; 17:267-78. [DOI: 10.3727/096368908784153850] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This study is a preliminary investigation to determine if bone marrow-derived cells, when implanted into freeze-injured urinary bladders, differentiate into smooth muscle cells and reconstruct smooth muscle layers. Bone marrow cells were harvested from femurs of male ICR mice and cultured in collagen-coated dishes for 7 days. After 5 days of culture, the cells were transfected with green fluorescent protein (GFP) genes for identification in recipient tissues. Three days prior to implantation, the posterior urinary bladder walls of female nude mice were injured with an iron bar refrigerated by dry ice. Seven days after the culture and 3 days after the injury, adherent, proliferating GFP-labeled bone marrow-derived cells (1.0 × 105 cells) were implanted into the injured regions. For controls, a cell-free solution was injected. At 14 days after implantation, the experimental urinary bladders were analyzed by histological, gene expression, and cystometric investigations. Just prior to implantation, the injured regions did not have any smooth muscle layers. After 14 days, the implanted cells surviving in the recipient tissues were detected with GFP antibody. The implanted regions had distinct smooth muscle layers composed of regenerated smooth muscle marker-positive cells. The implanted GFP-labeled cells differentiated into smooth muscle cells that formed into layers. The differentiated cells contacted each other within the implanted region as well as smooth muscle cells of the host. As a result, the reconstructed smooth muscle layers were integrated into the host tissues. Control mice injected with cell-free solution developed only few smooth muscle cells and no layers. Cystometric investigations showed that mice with implanted the cells developed bladder contractions similar to normal mice, whereas control mice did not. In summary, mouse bone marrow-derived cells can reconstruct layered smooth muscle structures in injured bladders to remediate urinary dysfunction.
Collapse
Affiliation(s)
- Tetsuya Imamura
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Yoshiaki Kinebuchi
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Osamu Ishizuka
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Satoshi Seki
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Yasuhiko Igawa
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Osamu Nishizawa
- Department of Urology, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| |
Collapse
|
27
|
van Laake LW, Passier R, Monshouwer-Kloots J, Verkleij AJ, Lips DJ, Freund C, den Ouden K, Ward-van Oostwaard D, Korving J, Tertoolen LG, van Echteld CJ, Doevendans PA, Mummery CL. Human embryonic stem cell-derived cardiomyocytes survive and mature in the mouse heart and transiently improve function after myocardial infarction. Stem Cell Res 2007; 1:9-24. [PMID: 19383383 DOI: 10.1016/j.scr.2007.06.001] [Citation(s) in RCA: 320] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 06/03/2007] [Indexed: 11/28/2022] Open
Abstract
Regeneration of the myocardium by transplantation of cardiomyocytes is an emerging therapeutic strategy. Human embryonic stem cells (HESC) form cardiomyocytes readily but until recently at low efficiency, so that preclinical studies on transplantation in animals are only just beginning. Here, we show the results of the first long-term (12 weeks) analysis of the fate of HESC-derived cardiomyocytes transplanted intramyocardially into healthy, immunocompromised (NOD-SCID) mice and in NOD-SCID mice that had undergone myocardial infarction (MI). Transplantation of mixed populations of differentiated HESC containing 20-25% cardiomyocytes in control mice resulted in rapid formation of grafts in which the cardiomyocytes became organized and matured over time and the noncardiomyocyte population was lost. Grafts also formed in mice that had undergone MI. Four weeks after transplantation and MI, this resulted in significant improvement in cardiac function measured by magnetic resonance imaging. However, at 12 weeks, this was not sustained despite graft survival. This suggested that graft size was still limiting despite maturation and organization of the transplanted cells. More generally, the results argued for requiring a minimum of 3 months follow-up in studies claiming to observe improved cardiac function, independent of whether HESC or other (adult) cell types are used for transplantation.
Collapse
Affiliation(s)
- Linda W van Laake
- Heart Lung Center, University Medical Center, GA Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Clément S, Stouffs M, Bettiol E, Kampf S, Krause KH, Chaponnier C, Jaconi M. Expression and function of alpha-smooth muscle actin during embryonic-stem-cell-derived cardiomyocyte differentiation. J Cell Sci 2006; 120:229-38. [PMID: 17179203 DOI: 10.1242/jcs.03340] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Three alpha-muscle actin isoforms are sequentially expressed during in vivo cardiac development. alpha-Smooth muscle actin is first and transiently expressed, followed by alpha-skeletal and finally alpha-cardiac actin. The significance of these transitions in actin gene expression during myogenesis remains to be determined. To understand whether actin isoforms have specific functions during cardiac development and cardiomyocyte contractility, we have hampered alpha-smooth muscle and alpha-skeletal actin expression and organization during embryonic stem cell differentiation towards cardiomyocyte. We show that the sequence of actin isoform expression displays similar pattern in the in vitro model and in mouse heart embryogenesis. Treatment with an interfering fusion peptide containing the N-terminal sequence of alpha-smooth muscle actin during a time window preceding spontaneous beating, prevents proper cardiac sarcomyogenesis, whereas alpha-skeletal actin-fusion peptide has no effect. Knockdown of alpha-smooth muscle actin in embryonic stem cells using RNA interference also affects cardiac differentiation. The application of both fusion peptides on beating embryoid bodies impairs frequency. These results suggest specific functional activities for actin isoforms in cardiogenesis and cardiomyocyte contractility.
Collapse
Affiliation(s)
- Sophie Clément
- Department of Geriatrics, Laboratory of Ageing, Geneva Hospital, Chêne-Bourg, Geneva, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Postnatally, heart muscle cells almost completely lose their ability to divide, which makes their loss after trauma irreversible. Potential repair by cell grafting or mobilizing endogenous cells is of particular interest for possible treatments for heart disease, where the poor capacity for cardiomyocyte proliferation probably contributes to the irreversibility of heart failure. Knowledge of the molecular mechanisms that underly formation of heart muscle cells might provide opportunities to repair the diseased heart by induction of (trans) differentiation of endogenous or exogenous cells into heart muscle cells. We briefly review the molecular mechanisms involved in early development of the linear heart tube by differentiation of mesodermal cells into heart muscle cells. Because the initial heart tube does not comprise all the cardiac compartments present in the adult heart, heart muscle cells are added to the distal borders of the tube and within the tube. At both distal borders, mesodermal cell are recruited into the cardiac lineage and, within the heart tube, muscular septa are formed. In this review, the relative late additions of heart muscle cells to the linear heart tube are described and the potential underlying molecular mechanisms are discussed.
Collapse
Affiliation(s)
- Maurice J B van den Hoff
- Molecular and Experimental Cardiology Group, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
30
|
Ehler E, Fowler VM, Perriard JC. Myofibrillogenesis in the developing chicken heart: Role of actin isoforms and of the pointed end actin capping protein tropomodulin during thin filament assembly. Dev Dyn 2004; 229:745-55. [PMID: 15042698 DOI: 10.1002/dvdy.10482] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Recently, important differences between myofibrillogenesis in cultured cardiomyocytes vs. the three-dimensional setting in situ could be determined. We investigated thin filament assembly in situ by confocal microscopy of whole-mount preparations of immunostained embryonic chicken hearts. Of interest, a distinct localisation of different actin isoforms was observed in immature thin filaments. Cardiac alpha-actin is restricted to filaments with a length comparable to mature thin filaments as soon as the first contractions occur, while vascular alpha-actin makes up filaments that extend toward the M-band. The pointed-end actin filament capping protein tropomodulin can be found initially in close association with the plasma membrane, but attains its mature localisation pattern at the ends of the thin filaments only comparatively late during myofibrillogenesis. Thus tropomodulin acts as a length stabilising element of actin filaments also in developing cardiomyocytes in situ, but plays an additional role together with membrane-associated actin filaments in the earliest steps of myofibril assembly.
Collapse
Affiliation(s)
- Elisabeth Ehler
- Institute of Cell Biology, ETH-Zürich Hönggerberg, Zürich, Switzerland
| | | | | |
Collapse
|
31
|
McMartin GA, Wirch E, Abraham N, Kargacin GJ. Postnatal changes in caldesmon expression and localization in cardiac myocytes. J Anat 2003; 203:369-77. [PMID: 14620377 PMCID: PMC1571179 DOI: 10.1046/j.1469-7580.2003.00228.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Caldesmon is a heat-stable protein found in both muscle and non-muscle tissue. It binds to a number of contractile and cytoskeletal proteins and may be involved in regulating acto-myosin interaction in smooth muscle cells and/or the assembly of microfilaments in muscle and non-muscle cells. We have shown previously that caldesmon is localized at the Z-lines in adult cardiac myocytes and that both the low- and high-molecular-weight forms (/-caldesmon and h-caldesmon, respectively) are present in atrial and ventricular myocytes. Here we examined the expression of caldesmon and its localization in freshly isolated cardiac myocytes during postnatal development and when these myocytes were grown in culture. We found that /-caldesmon is expressed in both neonatal and adult rat ventricular myocytes. The expression of h-caldesmon, however, was not detected in myocytes from newborn animals but increased during the first 2 weeks of postnatal development. Caldesmon was generally not co-localized with alpha-actinin at the Z-lines in neonatal myocytes but became increasingly more so during the first 2 weeks of postnatal development. When myocytes from 5- and 10-day-old rats were grown in primary culture, h-caldesmon expression decreased and caldesmon could not be detected at the Z-lines in the cultured cells. These results indicate that caldesmon plays a role at the Z-lines in adult cardiac myocytes; however, its localization at the Z-lines is not necessary for the prenatal development that occurs at these sites or for the establishment of a contractile phenotype in cultured cardiac myocytes.
Collapse
Affiliation(s)
- Gail A McMartin
- Department of Physiology and Biophysics, University of Calgary, Alberta, Canada
| | | | | | | |
Collapse
|
32
|
Kato H, Ohta S, Koshida S, Narita T, Taga T, Takeuchi Y, Sugita K. Expression of pericyte, mesangium and muscle markers in malignant rhabdoid tumor cell lines: differentiation-induction using 5-azacytidine. Cancer Sci 2003; 94:1059-65. [PMID: 14662021 PMCID: PMC11160295 DOI: 10.1111/j.1349-7006.2003.tb01401.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2003] [Revised: 10/10/2003] [Accepted: 10/11/2003] [Indexed: 12/22/2022] Open
Abstract
Malignant rhabdoid tumor (MRT) has been considered to have multiphenotypic diversity characteristics. Some MRTs exhibit a neural phenotype. However, it is still unclear whether MRT cells can display a skeletal muscle, smooth muscle or smooth muscle-like cell phenotype, like those of pericytes and mesangial cells. To determine if MRTs exhibit skeletal muscle cell or smooth muscle-like cell phenotypes, six MRT cell lines (TM87-16, STM91-01, TTC549, TTC642, YAM-RTK1 and TTC1240) were examined for markers of skeletal muscle (MyoD, myogenin, myf-5, myf-6, acetylcholine receptor-alpha, -beta and -gamma), smooth muscle (alpha-smooth muscle actin, SM-1 and SM22), and smooth muscle-like cells, such as pericytes (angiopoietin-1 and -2) and mesangial cells (megsin), using conventional RT-PCR, semi-quantitative PCR, western blotting and immunocytochemistry before and after differentiation-induction with 5-azacytidine. alpha-Smooth muscle actin and SM22 were detected in all six MRT cell lines, while MyoD and myf-5, crucial markers for skeletal myogenic determination, were not. The TM87-16 cell line expressed SM-1 and angiopoietin-1. TTC1240 also expressed angiopoietin-1. Interestingly, STM91-01 expressed megsin, a novel marker for mesangial cells, in addition to angiopoietin-1. Our results indicated that some MRTs exhibited smooth muscle and/or smooth muscle-like cell phenotypes and some renal MRTs might be of mesangial origin. Recently, smooth muscle and also smooth muscle-like cells have been considered to be of neuroectodermal origin. MRT can thus considered to belong to the category of primitive neuroectodermal tumors (PNETs) in the broad sense.
Collapse
Affiliation(s)
- Hirofumi Kato
- Department of Pediatrics, Shiga University of Medical Science, Tsukinowa, Ohtsu, Shiga 520-2192
| | | | | | | | | | | | | |
Collapse
|
33
|
Kruithof BPT, van den Hoff MJB, Wessels A, Moorman AFM. Cardiac muscle cell formation after development of the linear heart tube. Dev Dyn 2003; 227:1-13. [PMID: 12701094 DOI: 10.1002/dvdy.10269] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
After the development of the linear heart tube, additional myocardium is formed leading to the muscular mantle around the caval and pulmonary veins and the muscular septa in the embryonic heart. Here, we report the results of our in vivo and in vitro studies of this late myocardium-generating process in the mouse. By using an immunohistochemical approach, we determined that myocardium formation starts around embryonic day 12 in the dorsal mesocardium. In subsequent stages of development, the process extends downstream into the intracardiac mesenchymal tissues of the atrioventricular canal and outflow tract and upstream into the extracardiac mediastinal mesenchyme embedding the pulmonary and caval veins. Given the spatiotemporal pattern of myocardium formation, we applied a three-dimensional in vitro explant culture assay to investigate the myocardium-generating potential of the different cardiac compartments. We determined that this potential is stage- and mesenchyme-dependent. This latter finding suggests an important role for mesenchyme in myocardium formation after the development of the linear heart tube.
Collapse
Affiliation(s)
- Boudewijn P T Kruithof
- Experimental and Molecular Cardiology Group, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
34
|
Kruithof BPT, Van Den Hoff MJB, Tesink-Taekema S, Moorman AFM. Recruitment of intra- and extracardiac cells into the myocardial lineage during mouse development. THE ANATOMICAL RECORD. PART A, DISCOVERIES IN MOLECULAR, CELLULAR, AND EVOLUTIONARY BIOLOGY 2003; 271:303-14. [PMID: 12629673 DOI: 10.1002/ar.a.10033] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The tubular heart differentiates from the bilateral cardiac fields in the splanchnic mesoderm. The expression of smooth muscle proteins has been shown to accompany the early phases of cardiac muscle formation. In this study we show that during elongation of the arterial pole of the mouse linear heart tube, alpha-smooth muscle actin (alpha-Sma) expression extends in the area that has been shown to become recruited into the myocardial lineage, but does not yet express myocardial markers. These data suggest that alpha-Sma identifies mesodermal cells that during subsequent development will be recruited into the myocardial lineage. Myocardium formation is not only observed at the arterial pole, but also at the venous pole and in the intracardiac mesenchyme. This results in the formation of the caval and pulmonary myocardium, the smooth-walled atrial myocardium, the myocardial atrioventricular septum, and the myocardial outlet septum. To determine whether recruitment into the myocardial lineage also takes place in these regions, the spatiotemporal pattern of expression of alpha-Sma and of the myocardial markers sarcoplasmatic reticulum calcium ATPase (Serca2a), alpha-myosin heavy chain (Mhc), and beta-Mhc were examined. We show that prior to the expression of myocardial markers, alpha-Sma is expressed in these regions, which suggests that these mesodermal cells become recruited into the cardiac lineage after formation of the linear heart tube.
Collapse
Affiliation(s)
- Boudewijn P T Kruithof
- Experimental and Molecular Cardiology Group, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
35
|
Suurmeijer AJH, Clément S, Francesconi A, Bocchi L, Angelini A, Van Veldhuisen DJ, Spagnoli LG, Gabbiani G, Orlandi A. Alpha-actin isoform distribution in normal and failing human heart: a morphological, morphometric, and biochemical study. J Pathol 2003; 199:387-97. [PMID: 12579541 DOI: 10.1002/path.1311] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We investigated the distribution of alpha-skeletal, alpha-cardiac, and alpha-smooth muscle actin isoforms in human heart during development, hypertrophy, and failure. At 20 weeks of fetal life, alpha-skeletal actin was localized in a small proportion of subendocardial and papillary muscle cardiomyocytes. At this gestation time, diffuse alpha-cardiac actin staining was observed, associated with focal expression of alpha-smooth muscle actin. In normal adult subjects, alpha-skeletal actin positive cardiomyocytes were distributed in a transmural gradient with the highest proportion located subendocardially. In myocardial hypertrophy and cardiomyopathies, the amount of alpha-skeletal actin was increased and diffuse staining was seen in all layers of ventricular myocardium, with the exception of idiopathic dilated cardiomyopathies. Cardiomyocytes were negative for alpha-smooth muscle actin in all pathological situations studied. As expected, fibroblasts in post-infarct scars expressed alpha-smooth muscle actin and transforming growth factor-beta1 but, surprisingly, were negative for these proteins in interstitial fibrosis. Our results demonstrate that increased expression of alpha-skeletal actin in the diseased human heart is associated with increased myocyte stretch, increased wall stress, and pressure overload, but not with idiopathic dilated cardiomyopathies. They also suggest that fibrotic changes develop with different mechanisms in scars versus interstitial fibrosis.
Collapse
|
36
|
Miano JM. Mammalian smooth muscle differentiation: origins, markers and transcriptional control. Results Probl Cell Differ 2003; 38:39-59. [PMID: 12132398 DOI: 10.1007/978-3-540-45686-5_2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Joseph M Miano
- Center for Cardiovascular Research, Box 679, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, New York 14642, USA
| |
Collapse
|
37
|
Abstract
Heart morphogenesis comprises 2 major consecutive steps, viz. chamber formation followed by septation. Septation is the remodeling of the heart from a single-channel peristaltic pump to a dual-channel, synchronously contracting device with 1-way valves. In the human heart, septation occurs between 4 and 7 weeks of development. Cardiac looping and chamber formation bring the contributing structures into position to engage in septation. Cardiomyocytes that participate in chamber formation do not materially contribute to septation. The (re)discovery of the role of extracardiac mesenchymal tissue in atrioventricular septation, the appreciation that the formation of the right atrioventricular connection is more than a mere rightward expansion of the atrioventricular canal, the awareness that myocardium originating from the so-called anterior heart field regresses after its function as outflow-tract sphincter ceases, and the recent finding that the myocardialized proximal portion of the outflow-tract septum becomes the supraventricular crest have all significantly enhanced our understanding of the morphogenetic processes that contribute to septation. The bifurcation of the ventricular conduction system is the landmark that separates the contribution of the atrioventricular cushions and the outflow-tract ridges to septation and that divides the muscular ventricular septum in inlet, trabecular, and outlet portions.
Collapse
Affiliation(s)
- Wouter H Lamers
- Department of Anatomy and Embryology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|
38
|
Deruiter MC, Rensen SS, Coolen GP, Hierck BP, Bergwerff M, Debie WM, Gittenberger-De Groot AC, Van Eys GJ. Smoothelin expression during chicken embryogenesis: detection of an embryonic isoform. Dev Dyn 2001; 221:460-3. [PMID: 11500983 DOI: 10.1002/dvdy.1156] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Two isoforms of a novel smooth muscle cell (SMC) -specific cytoskeletal protein, smoothelin, have been described. In the adult chick, the 55-kDa smoothelin-A is expressed in visceral SMC, whereas the 120-kDa smoothelin-B is the major product in vascular SMC. Chicken was chosen to study smoothelin expression during embryogenesis and neonatally. Smoothelin-B was found in vascular SMC from stage 20 onward. In visceral SMC, smoothelin-B was present from stage 29 until hatching. Perinatally, a strong up-regulation of smoothelin synthesis was observed in visceral tissues, coinciding with a switch to the A-isoform. Transient smoothelin synthesis was observed in the somites and the developing heart. Western blotting revealed in these tissues a 62-kDa smoothelin isoform, designated smoothelin-C. Expression of the smoothelin isoforms seems to be strictly controlled with respect to cell type and developmental stage and may be related to the mode of contraction of the different cells.
Collapse
Affiliation(s)
- M C Deruiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Waller BR, McQuinn T, Phelps AL, Markwald RR, Lo CW, Thompson RP, Wessels A. Conotruncal anomalies in the trisomy 16 mouse: an immunohistochemical analysis with emphasis on the involvement of the neural crest. THE ANATOMICAL RECORD 2000; 260:279-93. [PMID: 11066038 DOI: 10.1002/1097-0185(20001101)260:3<279::aid-ar65>3.0.co;2-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The trisomy 16 (Ts16) mouse is generally considered a model for human Down's syndrome (trisomy 21). However, many of the cardiac defects in the Ts16 mouse do not reflect the heart malformations seen in patients suffering from this chromosomal disorder. In this study we describe the conotruncal malformations in mice with trisomy 16. The development of the outflow tract was immunohistochemically studied in serially sectioned hearts from 34 normal and 26 Ts16 mouse embryos ranging from 8.5 to 14.5 embryonic days. Conotruncal malformations observed in the Ts 16 embryos included double outlet right ventricle, persistent truncus arteriosus, Tetralogy of Fallot, and right-sided aortic arch. This spectrum of malformations is remarkably similar to that seen in humans suffering from DiGeorge syndrome (DGS). As perturbation of neural crest development has been proposed in the pathogenesis of DGS we specifically focussed on the fate of neural crest derived cells during outflow tract development of the Ts16 mouse using an antibody that enabled us to trace these cells during development. Severe perturbation of the neural crest-derived cell population was observed in each trisomic specimen. The abnormalities pertained to: 1) the size of the columns of neural crest-derived cells (or prongs); 2) the spatial orientation of these prongs within the mesenchymal tissues of the outflow tract; and 3) the location in which the neural crest cells interact with the myocardium. The latter abnormality appeared to be responsible for ectopic myocardialization found in trisomic embryos. Our observations strongly suggest that abnormal neural crest cell behavior is involved in the pathogenesis of the conotruncal malformations in the Ts16 mouse.
Collapse
Affiliation(s)
- B R Waller
- Department of Cell Biology & Anatomy, Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Vélez C, Aránega AE, Marchal JA, Melguizo C, Prados JC, Carrillo E, Aránega A. Development of chick cardiomyocytes: modulation of intermediate filaments by basic fibroblast and platelet-derived growth factors. Cells Tissues Organs 2000; 167:163-70. [PMID: 10971040 DOI: 10.1159/000016779] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Recent studies suggest that peptide growth factors play a functional role in cardiac muscle. To test whether embryonic cardiac muscle is a target for regulation by basic fibroblast growth factor and platelet-derived growth factor, we analyzed the effects of these peptides on the expression of the intermediate filaments desmin and vimentin at the subcellular level during development. Sodium dodecyl sulfate-gel electrophoresis, immunoblotting and fluorescence-activated cell sorting analysis were used to study the effect of basic fibroblast growth factor and platelet-derived growth factor on cultures of chick cardiomyocytes during development. Cytoplasmic and cytoskeletal concentrations of desmin and vimentin were dependent on the stage of embryonic development and on the type of growth factor added to the culture. The most significant finding was the increase in desmin expression in the cytoplasmic and cytoskeletal compartments after treatment with basic fibroblast growth factor (10 ng/ml) of chick heart cells at Hamburger and Hamilton stage 19. In more mature stages, basic fibroblast growth factor did not modify the levels of desmin expression. However, this factor led to a progressive deceleration in the rate of increase in vimentin expression. Platelet-derived growth factor increased vimentin expression in all stages studied, the greatest increases appearing in early stages of heart development. Our findings support the hypothesis that basic fibroblast growth factor plays a role in cardiomyocyte differentiation during the early stages of development, whereas platelet-derived growth factor has a dedifferentiating effect.
Collapse
Affiliation(s)
- C Vélez
- Department of Health Sciences, University of Almería, Spain
| | | | | | | | | | | | | |
Collapse
|
41
|
Colas JF, Lawson A, Schoenwolf GC. Evidence that translation of smooth muscle alpha-actin mRNA is delayed in the chick promyocardium until fusion of the bilateral heart-forming regions. Dev Dyn 2000; 218:316-30. [PMID: 10842359 DOI: 10.1002/(sici)1097-0177(200006)218:2<316::aid-dvdy6>3.0.co;2-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Heart development in the chick embryo proceeds from bilateral mesodermal primordia established during gastrulation. These primordia migrate to the midline and fuse into a single heart trough. During their migration as a cohesive sheet, the cells of the paired heart fields become epithelial and undergo cardiac differentiation, exhibiting organized myofibrils and rhythmic contractions near the time of their fusion. Between the stages of cardiomyoblast commitment and overt differentiation of cardiomyocytes, a significant time interval exists. Using a new riboprobe (usmaar) for whole-mount in situ hybridization in chick embryos, we report the earliest phases of smooth muscle alpha-actin (smaa) mRNA distribution during the precontractile developmental window. We show that ingressed heart-forming regions express smaa by the head-process stage (Hamburger and Hamilton stage 5). In addition, we used usmaar to study the formation and early morphogenesis of the heart. Consistent with fate mapping studies (Garcia-Martinez and Schoenwolf [1993] Dev. Biol. 159:706-719; Schoenwolf and Garcia-Martinez [1995] Cell Mol. Biol. Res. 41:233-240; Garcia-Martinez et al., in preparation), our results with this probe, combined with detailed histological and SEM analyses of the so-called cardiac crescent, demonstrate unequivocally that the heart arises from separated and paired heart rudiments, rather than from a single crescent-shaped rudiment (that is, prior to fusion of the paired heart rudiments to establish the straight-heart tube, the rostral midline of the cardiac crescent lacks mesodermal cells and consequently fails to label with usmaar). Smaa is also expressed in the splanchnic and somatic mesoderm, marking the earliest step in coelom formation. Consequently, we also used usmaar to describe formation of the pericardium. Finally, we provide evidence of a post-transcriptional level of control of smaa gene expression in the heart fields. Our results suggest that the expression of smaa may mark a primitive mesodermal state from which definitive cell types can be derived through inductive events.
Collapse
Affiliation(s)
- J F Colas
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City 84132, USA
| | | | | |
Collapse
|
42
|
Zammit PS, Kelly RG, Franco D, Brown N, Moorman AF, Buckingham ME. Suppression of atrial myosin gene expression occurs independently in the left and right ventricles of the developing mouse heart. Dev Dyn 2000; 217:75-85. [PMID: 10679931 DOI: 10.1002/(sici)1097-0177(200001)217:1<75::aid-dvdy7>3.0.co;2-l] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Many cardiac genes are broadly expressed in the early heart and become restricted to the atria or ventricles as development proceeds. Additional transcriptional differences between left and right compartments of the embryonic heart have been described recently, in particular for a number of transgenes containing cardiac regulatory elements. We now demonstrate that three myosin genes which become transcriptionally restricted to the atria between embryonic day (E) 12.5 and birth, alpha-myosin heavy chain (MHC), myosin light chain (MLC) 1A and MLC2A, are coordinately downregulated in the compact myocardium of the left ventricle before that of the right ventricle. alpha-MHC protein also accumulates in the right, but not left, compact ventricular myocardium during this period, suggesting that this transient regionalization contributes to fktal heart function. dHAND and eHAND, basic helix-loop-helix transcription factors known to be expressed in the right and left ventricles respectively at E10. 5, remain regionalized between E12.5 and E14.5. Downregulation of alpha-MHC, MLC1A, and MLC2A in iv/iv embryos, which have defective left/right patterning, initiates in the morphological left (systemic) ventricle regardless of its anatomical position on the right or left hand side of the heart. This points to the importance of left/right ventricular differences in sarcomeric gene expression patterns during fktal cardiogenesis and indicates that these differences originate in the embryo in response to anterior-posterior patterning of the heart tube rather than as a result of cardiac looping. Dev Dyn 2000;217:75-85.
Collapse
Affiliation(s)
- P S Zammit
- CNRS URA 1947, Département de Biologie Moléculaire, Institut Pasteur, Paris, France
| | | | | | | | | | | |
Collapse
|
43
|
Franco D, Markman MM, Wagenaar GT, Ya J, Lamers WH, Moorman AF. Myosin light chain 2a and 2v identifies the embryonic outflow tract myocardium in the developing rodent heart. Anat Rec (Hoboken) 1999; 254:135-46. [PMID: 9892427 DOI: 10.1002/(sici)1097-0185(19990101)254:1<135::aid-ar17>3.0.co;2-s] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The embryonic heart consists of five segments comprising the fast-conducting atrial and ventricular segments flanked by slow-conducting segments, i.e. inflow tract, atrioventricular canal and outflow tract. Although the incorporation of the flanking segments into the definitive atrial and ventricular chambers with development is generally accepted now, the contribution of the outflow tract myocardium to the definitive ventricles remained controversial mainly due to the lack of appropriate markers. For that reason we performed a detailed study of the pattern of expression of myosin light chain (MLC) 2a and 2v by in situ hybridization and immunohistochemistry during rat and mouse heart development. Expression of MLC2a mRNA displays a postero-anterior gradient in the tubular heart. In the embryonic heart it is down-regulated in the ventricular compartment and remains high in the outflow tract, atrioventricular canal, atria and inflow tract myocardium. MLC2v is strongly expressed in the ventricular myocardium and distinctly lower in the outflow tract and atrioventricular canal. The co-expression of MLC2a and MLC2v in the outflow tract and atrioventricular canal, together with the single expression in the atrial (MLC2a) and ventricular (MLC2v) myocardium, permits the delineation of their boundaries. With development, myocardial cells are observed in the lower endocardial ridges that share MLC2a and MLC2v expression with the myocardial cells of the outflow tract. In neonates, MLC2a continues to be expressed around both right and left semilunar valves, the outlet septum and the non-trabeculated right ventricular outlet. These findings demonstrate the contribution of the outflow tract to the definitive ventricles and demonstrate that the outlet septum is derived from outflow tract myocardium.
Collapse
Affiliation(s)
- D Franco
- Department of Anatomy and Embryology, Academic Medical Center, University of Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
44
|
Nakamura A, Isoyama S, Goto K. Vessel size-dependent expression of intermediate-sized filaments, calponin, and h-caldesmon in smooth muscle cells of human coronary arteries. Heart Vessels 1999; 14:253-61. [PMID: 10830922 DOI: 10.1007/bf01747855] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The arterial media is composed of a heterogeneous population of smooth muscle cells (SMCs). Recently, the properties of SMCs were observed to be heterogeneous not only among individual cells but also among arteries of the same vascular bed. To test the hypothesis that a site-specific heterogeneity exists in the SMCs of human coronary arteries, we examined the expression of desmin, vimentin, calponin, and high-molecular-weight (h-) caldesmon in arteries of various sizes. Specimens of arteries were obtained at autopsy from 12 patients: 6 adults (67 +/- 4 years old); 3 younger adults (26 +/- 2 years old); and 3 neonates. The size of the arteries was estimated by the number of SMC layers of the media. The expression was compared in SMCs of large arteries (>10 layers in adults, >5 layers in neonates), medium-sized arteries (5-10 layers in adults, 3-5 SMC layers in neonates), and small arteries (<3 layers). In adults, the percentage of arteries positive for desmin was lower in the small (17% +/- 3%) and medium-sized arteries (44% +/- 12%) than in the large arteries (94% +/- 6%) (P < 0.01). The percentage of arteries positive for calponin was also lower in the small (18% +/- 2%) and medium-sized arteries (66% +/- 5%) than in the large arteries (100%) (P < 0.01). The percentage for vimentin and h-caldesmon did not differ among large, medium-sized, and small arteries. These observations in adults were similar to those in younger adults or neonates. The phenotypes of medial SMCs are vessel size-dependent in human coronary arteries. This finding should be important for understanding the site-specific characteristics of vascular function in the regulation of myocardial perfusion or those of vascular responses to environmental changes.
Collapse
Affiliation(s)
- A Nakamura
- First Department of Internal Medicine, Tohoku University School of Medicine, Sendai, Japan
| | | | | |
Collapse
|
45
|
Ya J, Schilham MW, de Boer PA, Moorman AF, Clevers H, Lamers WH. Sox4-deficiency syndrome in mice is an animal model for common trunk. Circ Res 1998; 83:986-94. [PMID: 9815146 DOI: 10.1161/01.res.83.10.986] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Embryonic mice lacking functional Sox4 transcription factor die from cardiac failure at embryonic day (ED) 14. Heart morphogenesis in these embryos was analyzed in hematoxylin-azophlochsin or immunohistochemically stained, 3-dimensionally reconstructed serial sections between ED12 and ED14. Although Sox4 is expressed in the endocardially derived tissue of both the outflow tract and atrioventricular canal, Sox4-deficient hearts only suffer from defective transformation of the endocardial ridges into semilunar valves and from lack of fusion of these ridges, usually resulting in common trunk, although the least affected hearts should be classified as having a large infundibular septal defect. The more serious cases are, in addition, characterized by an abnormal number and position of the semilunar valve-leaflet anlagen, a configuration of the ridges typical for transposition of the great arteries (with linear rather than spiral course of both ridges and posterior position of the pulmonary trunk at the level of the valve), and variable size of the aorta relative to the pulmonary trunk. The coronary arteries always originated from the aorta, irrespective of its position relative to the pulmonary trunk. The restriction of the malformations to the arterial pole implies that the interaction between the endocardially derived tissue of the outflow tract and the neural crest-derived myofibroblasts determines proper development of the arterial pole.
Collapse
Affiliation(s)
- J Ya
- Department of Anatomy and Embryology, Academic Medical Center, University of Amsterdam, and the Department of Immunology, University of Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
46
|
Huang GY, Wessels A, Smith BR, Linask KK, Ewart JL, Lo CW. Alteration in connexin 43 gap junction gene dosage impairs conotruncal heart development. Dev Biol 1998; 198:32-44. [PMID: 9640330 DOI: 10.1006/dbio.1998.8891] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Connexin 43 (Cx43) knockout mice and transgenic mice (CMV43) overexpressing the Cx43 gap junction gene exhibit heart defects involving the conotruncus and right ventricle. Based on the heart phenotype and Cx43 gene and transgene expression pattern, we previously proposed that the heart defects may reflect a role for gap junctions in the modulation of cardiac neural crest development. To further elucidate the mechanism by which these heart defects may arise, fetal heart structure and function in these transgenic and knockout mice were examined by magnetic resonance microscopy and Doppler echocardiography. Magnetic resonance microscopy of E14.5 fetuses revealed an enlargement of the right ventricular chamber in the heterozygous Cx43 knockout and CMV43 transgenic mice. This was accompanied by thinning of the chamber wall. In the homozygous Cx43 knockout mouse, heart malformation was also restricted to the right ventricle. This was generally characterized by two pouches at the base of the pulmonary outflow tract, but occasionally hearts with a single pouch were found. Magnetic resonance microscopy showed in some of the CMV43 and Cx43 knockout mice an attenuation of the ductus arteriosus, a phenotype which may be indicative of outflow tract obstruction. This was confirmed by the in utero Doppler echocardiography, which showed increased outflow velocity in E12.5 to 14.5 CMV43 and Cx43 knockout fetuses. In some of these fetuses, Doppler analysis also revealed arrhythmia and absence of isovolemic contraction time. Further examination of these hearts by histology and immunohistochemistry showed abnormal myocardial development in the conotruncus. Particularly interesting was the presence of abundant subendocardial fibrous tissue expressing smooth muscle actin. In the developing heart, such mesenchyme in the outflow tract is usually considered neural crest-derived tissue. Together, these results confirm the importance of Cx43 gene dosage in conotruncal heart development and suggest that this likely involves a role for Cx43 gap junctions in cardiac crest development. In future studies, these transgenic mice may serve as valuable animal models for further studying the role of gap junctions and cardiac crest cells in conotruncal heart development.
Collapse
Affiliation(s)
- G Y Huang
- Department of Biology, Goddard Laboratory, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | | | |
Collapse
|
47
|
Affiliation(s)
- A F Moorman
- Cardiovascular Research Institute Amsterdam, Academic Medical Center, University of Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
48
|
Ya J, van den Hoff MJ, de Boer PA, Tesink-Taekema S, Franco D, Moorman AF, Lamers WH. Normal development of the outflow tract in the rat. Circ Res 1998; 82:464-72. [PMID: 9506707 DOI: 10.1161/01.res.82.4.464] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The outflow tract (OFT) provides the structural components forming the ventriculoarterial connection. The prevailing concept that this junction "rotates" to acquire its definitive topography also requires a concept of "counterrotation" and is difficult to reconcile with cell-marking studies. Rats between 10 embryonic days (EDs) and 2 postnatal days were stained immunohistochemically and by in situ hybridization. DNA replication was determined by incorporation of bromodeoxyuridine and apoptosis by the annexin V binding and terminal deoxynucleotidyl transferase-mediated dUTP-X nick end labeling (TUNEL) assays. Starting at ED12, cardiomyocytes in the distal (truncal) part of the OFT begin to shed their myocardial phenotype without proceeding into apoptosis, suggesting transdifferentiation. Myocardial regression is most pronounced on the dextroposterior side and continues until after birth, as revealed by the disappearance of the myocardial cuff surrounding the coronary roots and semilunar sinuses and by the establishment of fibrous continuity between mitral and aortic semilunar valves. Fusion of the endocardial ridges of the truncus on late ED13 is accompanied by the organization of alpha-smooth muscle actin-and nonmuscle myosin heavy chain-positive myofibroblasts into a central whorl and the appearance of the semilunar valve anlagen at their definitive topographical position within the proximal portion of the truncus. After fusion of the proximal (conal) portion of the endocardial ridges, many of the resident myofibroblasts undergo apoptosis and are replaced by cardiomyocytes. The distal myocardial boundary of the OFT is not a stable landmark but moves proximally over the spiraling course of the aortic and pulmonary routes, so that the semilunar valves develop at their definitive topographic position. After septation, the distal boundary of the OFT continues to regress, particularly in its subaortic portion. The myocardializing conus septum, on the other hand, becomes largely incorporated into the right ventricle. These opposite developments account for the pronounced asymmetry of the subaortic and subpulmonary outlets in the formed heart.
Collapse
Affiliation(s)
- J Ya
- Department of Anatomy and Embryology, Academic Medical Center, University of Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
49
|
Ya J, Erdtsieck-Ernste EB, de Boer PA, van Kempen MJ, Jongsma H, Gros D, Moorman AF, Lamers WH. Heart defects in connexin43-deficient mice. Circ Res 1998; 82:360-6. [PMID: 9486664 DOI: 10.1161/01.res.82.3.360] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiac malformation in connexin43 (CX43)-disrupted mice is restricted to the junction between right ventricle and outflow tract, even though CX43 is also expressed abundantly elsewhere. We analyzed cardiac morphogenesis in immunohistochemically and hybridohistochemically stained and three-dimensionally reconstructed serial sections of CX43-deficient embryos between embryonic day (ED) 10 and birth. The establishment of the D configuration in the ascending loop of CX43-deficient hearts is markedly retarded, so that the right ventricle retains a craniomedial position and is connected with the outflow tract by a more acute bend in ED10 and ED11 embryos. Because of the subsequent growth of the right ventricle, this condition usually evolves into a D loop, but when it persists, a "crisscross" configuration develops, with the atrioventricular cushions rotated 90 degrees, a horizontal muscular ventricular septum, and a parallel course of the endocardial ridges of the outflow tract. After ED12, large intertrabecular pouches develop at the ventricular side of both shelflike myocardial structures that support the endocardial ridges of the outflow tract, ie, at the location that was earlier characterized by the acute bend between the right ventricle and the outflow tract and that subsequently develops into the anterosuperior leaflet of the tricuspid valve. Retarded development of the D configuration in the ascending loop of the embryonic heart predisposes the myocardium at the junction of the right ventricle and outflow tract to excessive development of intertrabecular pouches during subsequent development.
Collapse
Affiliation(s)
- J Ya
- Department of Anatomy and Embryology, Academic Medical Center, University of Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|