1
|
Bønnelykke TH, Chabry MA, Perthame E, Dombrowsky G, Berger F, Dittrich S, Hitz MP, Desgrange A, Meilhac SM. Notch3 is an asymmetric gene and a modifier of heart looping defects in Nodal mouse mutants. PLoS Biol 2025; 23:e3002598. [PMID: 40163542 DOI: 10.1371/journal.pbio.3002598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/10/2025] [Accepted: 02/13/2025] [Indexed: 04/02/2025] Open
Abstract
The TGFβ secreted factor NODAL is a major left determinant required for the asymmetric morphogenesis of visceral organs, including the heart. Yet, when this signaling is absent, shape asymmetry, for example of the embryonic heart loop, is not fully abrogated, indicating that there are other factors regulating left-right patterning. Here, we used a tailored transcriptomic approach to screen for genes asymmetrically expressed in the field of heart progenitors. We thus identify Notch3 as a novel left-enriched gene and validate, by quantitative in situ hybridization, its transient asymmetry in the lateral plate mesoderm and node crown, overlapping with Nodal. In mutant embryos, we analyzed the regulatory hierarchy and demonstrate that Nodal in the lateral plate mesoderm amplifies Notch3 asymmetric expression. The function of Notch3 was uncovered in an allelic series of mutants. In single neonate mutants, we observe that Notch3 is required with partial penetrance for ventricle thickness, septation and aortic valve, in addition to its known role in coronary arteries. In compound mutants, we reveal that Notch3 acts as a genetic modifier of heart looping direction and shape defects in Nodal mutants. Whereas Notch3 was previously mainly associated with the CADASIL syndrome, our observations in the mouse and a human cohort support a novel role in congenital heart defects and laterality defects.
Collapse
Affiliation(s)
- Tobias Holm Bønnelykke
- Université Paris Cité, Imagine-Institut Pasteur Unit of Heart Morphogenesis , INSERM UMR1163, Paris, France
- Sorbonne Université, Collège Doctoral, Paris, France
| | - Marie-Amandine Chabry
- Université Paris Cité, Imagine-Institut Pasteur Unit of Heart Morphogenesis , INSERM UMR1163, Paris, France
| | - Emeline Perthame
- Université Paris Cité, Imagine-Institut Pasteur Unit of Heart Morphogenesis , INSERM UMR1163, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Gregor Dombrowsky
- Department for Medical Genetics, University of Oldenburg, Oldenburg, Germany
| | - Felix Berger
- Department of Congenital Heart Disease, Pediatric Cardiology Deutsches Herzzentrum der Charité, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sven Dittrich
- Department of Pediatric Cardiology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Marc-Phillip Hitz
- Department for Medical Genetics, University of Oldenburg, Oldenburg, Germany
- German Center for Cardiovascular Research (DZHK), Kiel, Germany
| | - Audrey Desgrange
- Université Paris Cité, Imagine-Institut Pasteur Unit of Heart Morphogenesis , INSERM UMR1163, Paris, France
| | - Sigolène M Meilhac
- Université Paris Cité, Imagine-Institut Pasteur Unit of Heart Morphogenesis , INSERM UMR1163, Paris, France
| |
Collapse
|
2
|
Gao H, Huang X, Chen W, Feng Z, Zhao Z, Li P, Tan C, Wang J, Zhuang Q, Gao Y, Min S, Yao Q, Qian M, Ma X, Wu F, Yan W, Sheng W, Huang G. Association of copy number variation in X chromosome-linked PNPLA4 with heterotaxy and congenital heart disease. Chin Med J (Engl) 2024; 137:1823-1834. [PMID: 38973237 PMCID: PMC12077557 DOI: 10.1097/cm9.0000000000003192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Heterotaxy (HTX) is a thoracoabdominal organ anomaly syndrome and commonly accompanied by congenital heart disease (CHD). The aim of this study was to analyze rare copy number variations (CNVs) in a HTX/CHD cohort and to examine the potential mechanisms contributing to HTX/CHD. METHODS Chromosome microarray analysis was used to identify rare CNVs in a cohort of 120 unrelated HTX/CHD patients, and available samples from parents were used to confirm the inheritance pattern. Potential candidate genes in CNVs region were prioritized via the DECIPHER database, and PNPLA4 was identified as the leading candidate gene. To validate, we generated PNPLA4 -overexpressing human induced pluripotent stem cell lines as well as pnpla4 -overexpressing zebrafish model, followed by a series of transcriptomic, biochemical and cellular analyses. RESULTS Seventeen rare CNVs were identified in 15 of the 120 HTX/CHD patients (12.5%). Xp22.31 duplication was one of the inherited CNVs identified in this HTX/CHD cohort, and PNPLA4 in the Xp22.31 was a candidate gene associated with HTX/CHD. PNPLA4 is expressed in the lateral plate mesoderm, which is known to be critical for left/right embryonic patterning as well as cardiomyocyte differentiation, and in the neural crest cell lineage. Through a series of in vivo and in vitro analyses at the molecular and cellular levels, we revealed that the biological function of PNPLA4 is importantly involved in the primary cilia formation and function via its regulation of energy metabolism and mitochondria-mediated ATP production. CONCLUSIONS Our findings demonstrated a significant association between CNVs and HTX/CHD. Our data strongly suggested that an increased genetic dose of PNPLA4 due to Xp22.31 duplication is a disease-causing risk factor for HTX/CHD.
Collapse
Affiliation(s)
- Han Gao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Xianghui Huang
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children’s Hospital, Xiamen, Fujian 361006, China
| | - Weicheng Chen
- Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Zhiyu Feng
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Zhengshan Zhao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Ping Li
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Chaozhong Tan
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Jinxin Wang
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Quannan Zhuang
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Yuan Gao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Shaojie Min
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Qinyu Yao
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Maoxiang Qian
- Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Xiaojing Ma
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
| | - Feizhen Wu
- Children’s Hospital of Fudan University, Shanghai 201102, China
| | - Weili Yan
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| | - Wei Sheng
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children’s Hospital, Xiamen, Fujian 361006, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| | - Guoying Huang
- Children’s Hospital of Fudan University, Shanghai 201102, China
- Shanghai Key Laboratory of Birth Defects, Shanghai 201102, China
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Children’s Hospital, Xiamen, Fujian 361006, China
- Research Unit of Early Intervention of Genetically Related Childhood Cardiovascular Diseases, Chinese Academy of Medical Sciences, Shanghai 201102, China
| |
Collapse
|
3
|
Katoh TA, Lange T, Nakajima Y, Yashiro K, Okada Y, Hamada H. BMP4 regulates asymmetric Pkd2 distribution in mouse nodal immotile cilia and ciliary mechanosensing required for left-right determination. Dev Dyn 2024. [PMID: 38984461 DOI: 10.1002/dvdy.727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/18/2024] [Accepted: 06/23/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Mouse nodal immotile cilia mechanically sense the bending direction for left-right (L-R) determination and activate the left-side-specific signaling cascade, leading to increased Nodal activity. Asymmetric distribution of Pkd2, a crucial channel for L-R determination, on immotile cilia has been reported recently. However, the causal relationship between the asymmetric Pkd2 distribution and direction-dependent flow sensing is not well understood. Furthermore, the underlying molecular mechanism directing this asymmetric Pkd2 distribution remains unclear. RESULTS The effects of several recombinant proteins and inhibitors on the Pkd2 distribution were analyzed using super-resolution microscopy. Notably, bone morphogenetic protein 4 (BMP4) affected the Pkd2 distribution. Additionally, three-dimensional manipulation of nodal immotile cilia using optical tweezers revealed that excess BMP4 caused defects in the mechanosensing ability of the cilia. CONCLUSIONS Experimental data together with model calculations suggest that BMP4 regulates the asymmetric distribution of Pkd2 in nodal immotile cilia, thereby affecting the ability of these cilia to sense the bending direction for L-R determination. This study, for the first time, provides insight into the relationship between the asymmetric protein distribution in cilia and their function.
Collapse
Affiliation(s)
- Takanobu A Katoh
- Department of Cell Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Organismal Patterning, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Tim Lange
- Laboratory for Organismal Patterning, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Yoshiro Nakajima
- Division of Anatomy and Developmental Biology, Department of Anatomy, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenta Yashiro
- Division of Anatomy and Developmental Biology, Department of Anatomy, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasushi Okada
- Department of Cell Biology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Cell Polarity Regulation, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Physics, Universal Biology Institute and International Research Center for Neurointelligence, The University of Tokyo, Hongo, Tokyo, Japan
| | - Hiroshi Hamada
- Laboratory for Organismal Patterning, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
4
|
Viswanathan PK, Chessel A, Molina MD, Haillot E, Lepage T. Maternal TGF-β ligand Panda breaks the radial symmetry of the sea urchin embryo by antagonizing the Nodal type II receptor ACVRII. PLoS Biol 2024; 22:e3002701. [PMID: 38913712 PMCID: PMC11239237 DOI: 10.1371/journal.pbio.3002701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 07/11/2024] [Accepted: 06/07/2024] [Indexed: 06/26/2024] Open
Abstract
In the highly regulative embryo of the sea urchin Paracentrotus lividus, establishment of the dorsal-ventral (D/V) axis critically depends on the zygotic expression of the TGF-β nodal in the ventral ectoderm. nodal expression is first induced ubiquitously in the 32-cell embryo and becomes progressively restricted to the presumptive ventral ectoderm by the early blastula stage. This early spatial restriction of nodal expression is independent of Lefty, and instead relies on the activity of Panda, a maternally expressed TGF-β ligand related to Lefty and Inhibins, which is required maternally for D/V axis specification. However, the mechanism by which Panda restricts the early nodal expression has remained enigmatic and it is not known if Panda works like a BMP ligand by opposing Nodal and antagonizing Smad2/3 signaling, or if it works like Lefty by sequestering an essential component of the Nodal signaling pathway. In this study, we report that Panda functions as an antagonist of the TGF-β type II receptor ACVRII (Activin receptor type II), which is the only type II receptor for Nodal signaling in the sea urchin and is also a type II receptor for BMP ligands. Inhibiting translation of acvrII mRNA disrupted D/V patterning across all 3 germ layers and caused acvrII morphants to develop with a typical Nodal loss-of-function phenotype. In contrast, embryos overexpressing acvrII displayed strong ectopic Smad1/5/8 signaling at blastula stages and developed as dorsalized larvae, a phenotype very similar to that caused by over activation of BMP signaling. Remarkably, embryos co-injected with acvrII mRNA and panda mRNA did not show ectopic Smad1/5/8 signaling and developed with a largely normal dorsal-ventral polarity. Furthermore, using an axis induction assay, we found that Panda blocks the ability of ACVRII to orient the D/V axis when overexpressed locally. Using co-immunoprecipitation, we showed that Panda physically interacts with ACVRII, as well as with the Nodal co-receptor Cripto, and with TBR3 (Betaglycan), which is a non-signaling receptor for Inhibins in mammals. At the molecular level, we have traced back the antagonistic activity of Panda to the presence of a single proline residue, conserved with all the Lefty factors, in the ACVRII binding motif of Panda, instead of a serine as in most of TGF-β ligands. Conversion of this proline to a serine converted Panda from an antagonist that opposed Nodal signaling and promoted dorsalization to an agonist that promoted Nodal signaling and triggered ventralization when overexpressed. Finally, using phylogenomics, we analyzed the emergence of the agonist and antagonist form of Panda in the course of evolution. Our data are consistent with the idea that the presence of a serine at that position, like in most TGF-β, was the ancestral condition and that the initial function of Panda was possibly in promoting and not in antagonizing Nodal signaling. These results highlight the existence of key functional and structural elements conserved between Panda and Lefty, allow to draw an intriguing parallel between sea urchin Panda and mammalian Inhibin α and raise the unexpected possibility that the original function of Panda may have been in activation of the Nodal pathway rather than in its inhibition.
Collapse
Affiliation(s)
| | - Aline Chessel
- Université Côte d’Azur, CNRS, Inserm, iBV, Nice, France
| | | | | | | |
Collapse
|
5
|
Gabriel GC, Lo CW. Molecular Pathways and Animal Models of Defects in Situs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:719-738. [PMID: 38884745 DOI: 10.1007/978-3-031-44087-8_43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Left-right patterning is among the least well understood of the three axes defining the body plan, and yet it is no less important, with left-right patterning defects causing structural birth defects with high morbidity and mortality, such as complex congenital heart disease, biliary atresia, or intestinal malrotation. The cell signaling pathways governing left-right asymmetry are highly conserved and involve multiple components of the TGF-β superfamily of cell signaling molecules. Central to left-right patterning is the differential activation of Nodal on the left, and BMP signaling on the right. In addition, a plethora of other cell signaling pathways including Shh, FGF, and Notch also contribute to the regulation of left-right patterning. In vertebrate embryos such as the mouse, frog, or zebrafish, the specification of left-right identity requires the left-right organizer (LRO) containing cells with motile and primary cilia that mediate the left-sided propagation of Nodal signaling, followed by left-sided activation of Lefty and then Pitx2, a transcription factor that specifies visceral organ asymmetry. While this overall scheme is well conserved, there are striking species differences, including the finding that motile cilia do not play a role in left-right patterning in some vertebrates. Surprisingly, the direction of heart looping, one of the first signs of organ left-right asymmetry, was recently shown to be specified by intrinsic cell chirality, not Nodal signaling, possibly a reflection of the early origin of Nodal signaling in radially symmetric organisms. How this intrinsic chirality interacts with downstream molecular pathways regulating visceral organ asymmetry will need to be further investigated to elucidate how disturbance in left-right patterning may contribute to complex CHD.
Collapse
Affiliation(s)
- George C Gabriel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Yagi H, Cui C, Saydmohammed M, Gabriel G, Baker C, Devine W, Wu Y, Lin JH, Malek M, Bais A, Murray S, Aronow B, Tsang M, Kostka D, Lo CW. Spatial transcriptome profiling uncovers metabolic regulation of left-right patterning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.21.537827. [PMID: 37131609 PMCID: PMC10153223 DOI: 10.1101/2023.04.21.537827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Left-right patterning disturbance can cause severe birth defects, but it remains least understood of the three body axes. We uncovered an unexpected role for metabolic regulation in left-right patterning. Analysis of the first spatial transcriptome profile of left-right patterning revealed global activation of glycolysis, accompanied by right-sided expression of Bmp7 and genes regulating insulin growth factor signaling. Cardiomyocyte differentiation was left-biased, which may underlie the specification of heart looping orientation. This is consistent with known Bmp7 stimulation of glycolysis and glycolysis suppression of cardiomyocyte differentiation. Liver/lung laterality may be specified via similar metabolic regulation of endoderm differentiation. Myo1d , found to be left-sided, was shown to regulate gut looping in mice, zebrafish, and human. Together these findings indicate metabolic regulation of left-right patterning. This could underlie high incidence of heterotaxy-related birth defects in maternal diabetes, and the association of PFKP, allosteric enzyme regulating glycolysis, with heterotaxy. This transcriptome dataset will be invaluable for interrogating birth defects involving laterality disturbance.
Collapse
|
7
|
Saba TG, Geddes GC, Ware SM, Schidlow DN, Del Nido PJ, Rubalcava NS, Gadepalli SK, Stillwell T, Griffiths A, Bennett Murphy LM, Barber AT, Leigh MW, Sabin N, Shapiro AJ. A multi-disciplinary, comprehensive approach to management of children with heterotaxy. Orphanet J Rare Dis 2022; 17:351. [PMID: 36085154 PMCID: PMC9463860 DOI: 10.1186/s13023-022-02515-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/04/2022] [Indexed: 11/10/2022] Open
Abstract
Heterotaxy (HTX) is a rare condition of abnormal thoraco-abdominal organ arrangement across the left-right axis of the body. The pathogenesis of HTX includes a derangement of the complex signaling at the left-right organizer early in embryogenesis involving motile and non-motile cilia. It can be inherited as a single-gene disorder, a phenotypic feature of a known genetic syndrome or without any clear genetic etiology. Most patients with HTX have complex cardiovascular malformations requiring surgical intervention. Surgical risks are relatively high due to several serious comorbidities often seen in patients with HTX. Asplenia or functional hyposplenism significantly increase the risk for sepsis and therefore require antimicrobial prophylaxis and immediate medical attention with fever. Intestinal rotation abnormalities are common among patients with HTX, although volvulus is rare and surgical correction carries substantial risk. While routine screening for intestinal malrotation is not recommended, providers and families should promptly address symptoms concerning for volvulus and biliary atresia, another serious morbidity more common among patients with HTX. Many patients with HTX have chronic lung disease and should be screened for primary ciliary dyskinesia, a condition of respiratory cilia impairment leading to bronchiectasis. Mental health and neurodevelopmental conditions need to be carefully considered among this population of patients living with a substantial medical burden. Optimal care of children with HTX requires a cohesive team of primary care providers and experienced subspecialists collaborating to provide compassionate, standardized and evidence-based care. In this statement, subspecialty experts experienced in HTX care and research collaborated to provide expert- and evidence-based suggestions addressing the numerous medical issues affecting children living with HTX.
Collapse
Affiliation(s)
- Thomas G Saba
- Department of Pediatrics, Pulmonary Division, University of Michigan Medical School, 1500 E. Medical Center Drive, Ann Arbor, MI, USA.
| | - Gabrielle C Geddes
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephanie M Ware
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David N Schidlow
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro J Del Nido
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nathan S Rubalcava
- Department of Surgery, Section of Pediatric Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Samir K Gadepalli
- Department of Surgery, Section of Pediatric Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Terri Stillwell
- Department of Pediatrics, Infectious Disease Division, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anne Griffiths
- Department of Pediatrics, Pulmonary/Critical Care Division, Children's Minnesota and Children's Respiratory and Critical Care Specialists, Minneapolis, MN, USA
| | - Laura M Bennett Murphy
- Department of Pediatrics, Division of Pediatric Psychiatry and Behavioral Health, University of Utah, Primary Children's Hospital, Salt Lake City, UT, USA
| | - Andrew T Barber
- Department of Pediatrics, Division of Pulmonology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Margaret W Leigh
- Department of Pediatrics, Division of Pulmonology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Necia Sabin
- Heterotaxy Connection, Eagle Mountain, UT, USA
| | - Adam J Shapiro
- Department of Pediatrics, McGill University Health Centre Research Institute, Montreal, QC, Canada
| |
Collapse
|
8
|
Hill CS. Establishment and interpretation of NODAL and BMP signaling gradients in early vertebrate development. Curr Top Dev Biol 2022; 149:311-340. [PMID: 35606059 DOI: 10.1016/bs.ctdb.2021.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Transforming growth factor β (TGF-β) family ligands play crucial roles in orchestrating early embryonic development. Most significantly, two family members, NODAL and BMP form signaling gradients and indeed in fish, frogs and sea urchins these two opposing gradients are sufficient to organize a complete embryonic axis. This review focuses on how these gradients are established and interpreted during early vertebrate development. The review highlights key principles that are emerging, in particular the importance of signaling duration as well as ligand concentration in both gradient generation and their interpretation. Feedforward and feedback loops involving other signaling pathways are also essential for providing spatial and temporal information downstream of the NODAL and BMP signaling pathways. Finally, new data suggest the existence of buffering mechanisms, whereby early signaling defects can be readily corrected downstream later in development, suggesting that signaling gradients do not have to be as precise as previously thought.
Collapse
Affiliation(s)
- Caroline S Hill
- Developmental Signalling Laboratory, The Francis Crick Institute, London, United Kingdom.
| |
Collapse
|
9
|
Little RB, Norris DP. Right, left and cilia: How asymmetry is established. Semin Cell Dev Biol 2021; 110:11-18. [PMID: 32571625 DOI: 10.1016/j.semcdb.2020.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
Abstract
The initial breaking of left-right (L-R) symmetry in the embryo is controlled by a motile-cilia-driven leftward fluid flow in the left-right organiser (LRO), resulting in L-R asymmetric gene expression flanking the LRO. Ultimately this results in left- but not right-sided activation of the Nodal-Pitx2 pathway in more lateral tissues. While aspects of the initial breaking event clearly vary between vertebrates, events in the Lateral Plate Mesoderm (LPM) are conserved through the vertebrate lineage. Evidence from model systems and humans highlights the role of cilia both in the initial symmetry breaking and in the ability of more lateral tissues to exhibit asymmetric gene expression. In this review we concentrate on the process of L-R determination in mouse and humans.
Collapse
Affiliation(s)
- Rosie B Little
- MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Dominic P Norris
- MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK.
| |
Collapse
|
10
|
Wang W, Zheng X, Song H, Yang J, Liu X, Wang Y, Zhang M, Zhang Z. Spatial and temporal deletion reveals a latent effect of Megf8 on the left-right patterning and heart development. Differentiation 2020; 113:19-25. [PMID: 32203821 DOI: 10.1016/j.diff.2020.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 01/23/2023]
Abstract
Laterality disease is frequently associated with congenital heart disease (CHD). However, it is unclear what is behind this association, a pleiotropic effect of common genetic causes of laterality diseases or the impact of abnormal left-right patterning on the downstream cardiovascular development. MEGF8 is a disease gene of Carpenter syndrome characterized by defective lateralization and CHD. Here we performed spatial and temporal deletion to dissect the tissue and time requirements of Megf8 on cardiovascular development. None of conditional deletions in cardiomyocytes, endothelium/endocardium, epicardium, cardiac mesoderm or neural crest cells led to cardiovascular defects. More surprisingly, temporal deletion with a ubiquitous Cre driver at embryonic day 7.5 (E7.5), a time point before symmetry break and cardiogenesis, causes preaxial polydactyly (PPD) and exencephaly, but not laterality and cardiovascular defects. These data suggested that Megf8 was dispensable for cardiac organogenesis. Only with E6.5 deletion, we observed aortic arch artery defects including right aortic arch, an indicator of reversed left-right patterning. The concurrence of laterality and cardiovascular defects in pre-streak stage deletion rather than cardiac organogenesis stage deletion indicates that the laterality defect may directly impact heart development. Interestingly, the latent effect of Megf8 on the left-right patterning suggests that the regulation of laterality may be much earlier than we previously thought.
Collapse
Affiliation(s)
- Wenfeng Wang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiaoling Zheng
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hejie Song
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Junjie Yang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiangyang Liu
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ye Wang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Min Zhang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zhen Zhang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
11
|
Yamak A, Hu D, Mittal N, Buikema JW, Ditta S, Lutz PG, Moog-Lutz C, Ellinor PT, Domian IJ. Loss of Asb2 Impairs Cardiomyocyte Differentiation and Leads to Congenital Double Outlet Right Ventricle. iScience 2020; 23:100959. [PMID: 32179481 PMCID: PMC7078385 DOI: 10.1016/j.isci.2020.100959] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/17/2019] [Accepted: 02/26/2020] [Indexed: 11/21/2022] Open
Abstract
Defining the pathways that control cardiac development facilitates understanding the pathogenesis of congenital heart disease. Herein, we identify enrichment of a Cullin5 Ub ligase key subunit, Asb2, in myocardial progenitors and differentiated cardiomyocytes. Using two conditional murine knockouts, Nkx+/Cre.Asb2fl/fl and AHF-Cre.Asb2fl/fl, and tissue clarifying technique, we reveal Asb2 requirement for embryonic survival and complete heart looping. Deletion of Asb2 results in upregulation of its target Filamin A (Flna), and concurrent Flna deletion partially rescues embryonic lethality. Conditional AHF-Cre.Asb2 knockouts harboring one Flna allele have double outlet right ventricle (DORV), which is rescued by biallelic Flna excision. Transcriptomic and immunofluorescence analyses identify Tgfβ/Smad as downstream targets of Asb2/Flna. Finally, using CRISPR/Cas9 genome editing, we demonstrate Asb2 requirement for human cardiomyocyte differentiation suggesting a conserved mechanism between mice and humans. Collectively, our study provides deeper mechanistic understanding of the role of the ubiquitin proteasome system in cardiac development and suggests a previously unidentified murine model for DORV. Flna removal partially rescues embryonic lethality of Asb2-heart-specific knockout AHF-Asb2 knockouts harboring one Flna allele have double outlet right ventricle Asb2-Flna regulate TGFβ-Smad2 signaling in the heart Conserved role of Asb2 in heart morphogenesis between mice and humans
Collapse
Affiliation(s)
- Abir Yamak
- Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Dongjian Hu
- Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Nikhil Mittal
- Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA
| | - Jan W Buikema
- Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; University Medical Center Utrecht, 3584 CX Utrecht, Netherlands
| | - Sheraz Ditta
- Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; Department of Pharmaceutical Sciences, Utrecht University, 3512 JE Utrecht, Netherlands
| | - Pierre G Lutz
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Christel Moog-Lutz
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Patrick T Ellinor
- Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ibrahim J Domian
- Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
12
|
Sempou E, Khokha MK. Genes and mechanisms of heterotaxy: patients drive the search. Curr Opin Genet Dev 2019; 56:34-40. [PMID: 31234044 DOI: 10.1016/j.gde.2019.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/03/2019] [Accepted: 05/11/2019] [Indexed: 12/17/2022]
Abstract
Heterotaxy, a disorder in which visceral organs, including the heart, are mispatterned along the left-right body axis, contributes to particularly severe forms of congenital heart disease that are difficult to mitigate even despite surgical advances. A higher incidence of heterotaxy among individuals with blood kinship and the existence of rare monogenic disease forms suggest the existence of a genetic component, but the genetic and phenotypic heterogeneity of the disease have rendered gene discovery challenging. Next generation genomics in patients with syndromic, but also non-syndromic and sporadic heterotaxy, have recently helped to uncover new candidate disease genes, expanding the pool of genes already identified via traditional animal studies. Further characterization of these new genes in animal models has uncovered fascinating mechanisms of left-right axis development. In this review, we will discuss recent findings on the functions of heterotaxy genes with identified patient alleles.
Collapse
Affiliation(s)
- Emily Sempou
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, United States.
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, United States
| |
Collapse
|
13
|
Desgrange A, Le Garrec JF, Meilhac SM. Left-right asymmetry in heart development and disease: forming the right loop. Development 2018; 145:145/22/dev162776. [PMID: 30467108 DOI: 10.1242/dev.162776] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Extensive studies have shown how bilateral symmetry of the vertebrate embryo is broken during early development, resulting in a molecular left-right bias in the mesoderm. However, how this early asymmetry drives the asymmetric morphogenesis of visceral organs remains poorly understood. The heart provides a striking model of left-right asymmetric morphogenesis, undergoing rightward looping to shape an initially linear heart tube and align cardiac chambers. Importantly, abnormal left-right patterning is associated with severe congenital heart defects, as exemplified in heterotaxy syndrome. Here, we compare the mechanisms underlying the rightward looping of the heart tube in fish, chick and mouse embryos. We propose that heart looping is not only a question of direction, but also one of fine-tuning shape. This is discussed in the context of evolutionary and clinical perspectives.
Collapse
Affiliation(s)
- Audrey Desgrange
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France.,INSERM UMR1163, Université Paris Descartes, 75015 Paris, France
| | - Jean-François Le Garrec
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France.,INSERM UMR1163, Université Paris Descartes, 75015 Paris, France
| | - Sigolène M Meilhac
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France .,INSERM UMR1163, Université Paris Descartes, 75015 Paris, France
| |
Collapse
|
14
|
The Drosophila homologue of MEGF8 is essential for early development. Sci Rep 2018; 8:8790. [PMID: 29884872 PMCID: PMC5993795 DOI: 10.1038/s41598-018-27076-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/23/2018] [Indexed: 12/15/2022] Open
Abstract
Mutations of the gene MEGF8 cause Carpenter syndrome in humans, and the mouse orthologue has been functionally associated with Nodal and Bmp4 signalling. Here, we have investigated the phenotype associated with loss-of-function of CG7466, a gene that encodes the Drosophila homologue of MEGF8. We generated three different frame-shift null mutations in CG7466 using CRISPR/Cas9 gene editing. Heterozygous flies appeared normal, but homozygous animals had disorganised denticle belts and died as 2nd or 3rd instar larvae. Larvae were delayed in transition to 3rd instars and showed arrested growth, which was associated with abnormal feeding behaviour and prolonged survival when yeast food was supplemented with sucrose. RNAi-mediated knockdown using the Gal4-UAS system resulted in lethality with ubiquitous and tissue-specific Gal4 drivers, and growth defects including abnormal bristle number and orientation in a subset of escapers. We conclude that CG7466 is essential for larval development and that diminished function perturbs denticle and bristle formation.
Collapse
|
15
|
Abstract
TGF-β family ligands function in inducing and patterning many tissues of the early vertebrate embryonic body plan. Nodal signaling is essential for the specification of mesendodermal tissues and the concurrent cellular movements of gastrulation. Bone morphogenetic protein (BMP) signaling patterns tissues along the dorsal-ventral axis and simultaneously directs the cell movements of convergence and extension. After gastrulation, a second wave of Nodal signaling breaks the symmetry between the left and right sides of the embryo. During these processes, elaborate regulatory feedback between TGF-β ligands and their antagonists direct the proper specification and patterning of embryonic tissues. In this review, we summarize the current knowledge of the function and regulation of TGF-β family signaling in these processes. Although we cover principles that are involved in the development of all vertebrate embryos, we focus specifically on three popular model organisms: the mouse Mus musculus, the African clawed frog of the genus Xenopus, and the zebrafish Danio rerio, highlighting the similarities and differences between these species.
Collapse
Affiliation(s)
- Joseph Zinski
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Benjamin Tajer
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Mary C Mullins
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| |
Collapse
|
16
|
Soukup V, Kozmik Z. The Bmp signaling pathway regulates development of left-right asymmetry in amphioxus. Dev Biol 2018; 434:164-174. [PMID: 29224891 DOI: 10.1016/j.ydbio.2017.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 12/05/2017] [Accepted: 12/05/2017] [Indexed: 01/31/2023]
Abstract
Establishment of asymmetry along the left-right (LR) body axis in vertebrates requires interplay between Nodal and Bmp signaling pathways. In the basal chordate amphioxus, the left-sided activity of the Nodal signaling has been attributed to the asymmetric morphogenesis of paraxial structures and pharyngeal organs, however the role of Bmp signaling in LR asymmetry establishment has not been addressed to date. Here, we show that Bmp signaling is necessary for the development of LR asymmetric morphogenesis of amphioxus larvae through regulation of Nodal signaling. Loss of Bmp signaling results in loss of the left-sided expression of Nodal, Gdf1/3, Lefty and Pitx and in gain of ectopic expression of Cerberus on the left side. As a consequence, the larvae display loss of the offset arrangement of axial structures, loss of the left-sided pharyngeal organs including the mouth, and ectopic development of the right-sided organs on the left side. Bmp inhibition thus phenocopies inhibition of Nodal signaling and results in the right isomerism. We conclude that Bmp and Nodal pathways act in concert to specify the left side and that Bmp signaling plays a fundamental role during LR development in amphioxus.
Collapse
Affiliation(s)
- Vladimir Soukup
- Institute of Molecular Genetics, The Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic.
| | - Zbynek Kozmik
- Institute of Molecular Genetics, The Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic
| |
Collapse
|
17
|
Liu C, Peng G, Jing N. TGF-β signaling pathway in early mouse development and embryonic stem cells. Acta Biochim Biophys Sin (Shanghai) 2018; 50:68-73. [PMID: 29190317 DOI: 10.1093/abbs/gmx120] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/31/2017] [Indexed: 12/30/2022] Open
Abstract
TGF-β superfamily signaling pathways essentially contribute to the broad spectrum of early developmental events including embryonic patterning, cell fate determination and dynamic movements. In this review, we first introduced some key developmental processes that require TGF-β signaling to show the fundamental importance of these pathways. Then we discuss how their activities are regulated, and new findings about how the TGF-β superfamily ligands bind to the chromatin to regulate transcription during embryo development.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Guangdun Peng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
18
|
Boghossian NS, Sicko RJ, Giannakou A, Dimopoulos A, Caggana M, Tsai MY, Yeung EH, Pankratz N, Cole BR, Romitti PA, Browne ML, Fan R, Liu A, Kay DM, Mills JL. Rare copy number variants identified in prune belly syndrome. Eur J Med Genet 2017; 61:145-151. [PMID: 29174092 DOI: 10.1016/j.ejmg.2017.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/31/2017] [Accepted: 11/21/2017] [Indexed: 11/26/2022]
Abstract
Prune belly syndrome (PBS), also known as Eagle-Barrett syndrome, is a rare congenital disorder characterized by absence or hypoplasia of the abdominal wall musculature, urinary tract anomalies, and cryptorchidism in males. The etiology of PBS is largely unresolved, but genetic factors are implicated given its recurrence in families. We examined cases of PBS to identify novel pathogenic copy number variants (CNVs). A total of 34 cases (30 males and 4 females) with PBS identified from all live births in New York State (1998-2005) were genotyped using Illumina HumanOmni2.5 microarrays. CNVs were prioritized if they were absent from in-house controls, encompassed ≥10 consecutive probes, were ≥20 Kb in size, had ≤20% overlap with common variants in population reference controls, and had ≤20% overlap with any variant previously detected in other birth defect phenotypes screened in our laboratory. We identified 17 candidate autosomal CNVs; 10 cases each had one CNV and four cases each had two CNVs. The CNVs included a 158 Kb duplication at 4q22 that overlaps the BMPR1B gene; duplications of different sizes carried by two cases in the intron of STIM1 gene; a 67 Kb duplication 202 Kb downstream of the NOG gene, and a 1.34 Mb deletion including the MYOCD gene. The identified rare CNVs spanned genes involved in mesodermal, muscle, and urinary tract development and differentiation, which might help in elucidating the genetic contribution to PBS. We did not have parental DNA and cannot identify whether these CNVs were de novo or inherited. Further research on these CNVs, particularly BMP signaling is warranted to elucidate the pathogenesis of PBS.
Collapse
Affiliation(s)
- Nansi S Boghossian
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, United States; Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States.
| | - Robert J Sicko
- Division of Genetics, Wadsworth Center, Department of Health, Albany, NY, United States
| | - Andreas Giannakou
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Aggeliki Dimopoulos
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Michele Caggana
- Division of Genetics, Wadsworth Center, Department of Health, Albany, NY, United States
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Edwina H Yeung
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Benjamin R Cole
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Paul A Romitti
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, IA, United States
| | - Marilyn L Browne
- New York State Department of Health, Congenital Malformations Registry, Albany, NY, United States; University at Albany School of Public Health, Rensselaer, NY, United States
| | - Ruzong Fan
- Department of Biostatistics, Bioinformatics, and Biomathematics, Georgetown University Medical Center (GUMC), Washington, DC, United States
| | - Aiyi Liu
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Denise M Kay
- Division of Genetics, Wadsworth Center, Department of Health, Albany, NY, United States
| | - James L Mills
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
19
|
Lin CY, Tsai MY, Liu YH, Lu YF, Chen YC, Lai YR, Liao HC, Lien HW, Yang CH, Huang CJ, Hwang SPL. Klf8 regulates left-right asymmetric patterning through modulation of Kupffer's vesicle morphogenesis and spaw expression. J Biomed Sci 2017; 24:45. [PMID: 28716076 PMCID: PMC5513281 DOI: 10.1186/s12929-017-0351-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/07/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Although vertebrates are bilaterally symmetric organisms, their internal organs are distributed asymmetrically along a left-right axis. Disruption of left-right axis asymmetric patterning often occurs in human genetic disorders. In zebrafish embryos, Kupffer's vesicle, like the mouse node, breaks symmetry by inducing asymmetric expression of the Nodal-related gene, spaw, in the left lateral plate mesoderm (LPM). Spaw then stimulates transcription of itself and downstream genes, including lft1, lft2, and pitx2, specifically in the left side of the diencephalon, heart and LPM. This developmental step is essential to establish subsequent asymmetric organ positioning. In this study, we evaluated the role of krüppel-like factor 8 (klf8) in regulating left-right asymmetric patterning in zebrafish embryos. METHODS Zebrafish klf8 expression was disrupted by both morpholino antisense oligomer-mediated knockdown and a CRISPR-Cas9 system. Whole-mount in situ hybridization was conducted to evaluate gene expression patterns of Nodal signalling components and the positions of heart and visceral organs. Dorsal forerunner cell number was evaluated in Tg(sox17:gfp) embryos and the length and number of cilia in Kupffer's vesicle were analyzed by immunocytochemistry using an acetylated tubulin antibody. RESULTS Heart jogging, looping and visceral organ positioning were all defective in zebrafish klf8 morphants. At the 18-22 s stages, klf8 morphants showed reduced expression of genes encoding Nodal signalling components (spaw, lft1, lft2, and pitx2) in the left LPM, diencephalon, and heart. Co-injection of klf8 mRNA with klf8 morpholino partially rescued spaw expression. Furthermore, klf8 but not klf8△zf overexpressing embryos showed dysregulated bilateral expression of Nodal signalling components at late somite stages. At the 10s stage, klf8 morphants exhibited reductions in length and number of cilia in Kupffer's vesicle, while at 75% epiboly, fewer dorsal forerunner cells were observed. Interestingly, klf8 mutant embryos, generated by a CRISPR-Cas9 system, showed bilateral spaw expression in the LPM at late somite stages. This observation may be partly attributed to compensatory upregulation of klf12b, because klf12b knockdown reduced the percentage of klf8 mutants exhibiting bilateral spaw expression. CONCLUSIONS Our results demonstrate that zebrafish Klf8 regulates left-right asymmetric patterning by modulating both Kupffer's vesicle morphogenesis and spaw expression in the left LPM.
Collapse
Affiliation(s)
- Che-Yi Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan.,Present address: Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Ming-Yuan Tsai
- Graduate Institute of Life Sciences, National Defence Medical Center, National Defence University, Neihu, Taipei, Taiwan.,Present address: Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yu-Hsiu Liu
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Fen Lu
- Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei, 11529, Taiwan
| | - Yi-Chung Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei, 11529, Taiwan
| | - Yun-Ren Lai
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan
| | - Hsin-Chi Liao
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan
| | - Huang-Wei Lien
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | | | - Chang-Jen Huang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Sheng-Ping L Hwang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan. .,Department of Life Science, National Taiwan University, Taipei, Taiwan. .,Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei, 11529, Taiwan.
| |
Collapse
|
20
|
Weaver CA, Miller SF, da Fontoura CSG, Wehby GL, Amendt BA, Holton NE, Allareddy V, Southard TE, Moreno Uribe LM. Candidate gene analyses of 3-dimensional dentoalveolar phenotypes in subjects with malocclusion. Am J Orthod Dentofacial Orthop 2017; 151:539-558. [PMID: 28257739 DOI: 10.1016/j.ajodo.2016.08.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 12/01/2022]
Abstract
INTRODUCTION Genetic studies of malocclusion etiology have identified 4 deleterious mutations in genes DUSP6,ARHGAP21, FGF23, and ADAMTS1 in familial Class III cases. Although these variants may have large impacts on Class III phenotypic expression, their low frequency (<1%) makes them unlikely to explain most malocclusions. Thus, much of the genetic variation underlying the dentofacial phenotypic variation associated with malocclusion remains unknown. In this study, we evaluated associations between common genetic variations in craniofacial candidate genes and 3-dimensional dentoalveolar phenotypes in patients with malocclusion. METHODS Pretreatment dental casts or cone-beam computed tomographic images from 300 healthy subjects were digitized with 48 landmarks. The 3-dimensional coordinate data were submitted to a geometric morphometric approach along with principal component analysis to generate continuous phenotypes including symmetric and asymmetric components of dentoalveolar shape variation, fluctuating asymmetry, and size. The subjects were genotyped for 222 single-nucleotide polymorphisms in 82 genes/loci, and phenotpye-genotype associations were tested via multivariate linear regression. RESULTS Principal component analysis of symmetric variation identified 4 components that explained 68% of the total variance and depicted anteroposterior, vertical, and transverse dentoalveolar discrepancies. Suggestive associations (P < 0.05) were identified with PITX2, SNAI3, 11q22.2-q22.3, 4p16.1, ISL1, and FGF8. Principal component analysis for asymmetric variations identified 4 components that explained 51% of the total variations and captured left-to-right discrepancies resulting in midline deviations, unilateral crossbites, and ectopic eruptions. Suggestive associations were found with TBX1AJUBA, SNAI3SATB2, TP63, and 1p22.1. Fluctuating asymmetry was associated with BMP3 and LATS1. Associations for SATB2 and BMP3 with asymmetric variations remained significant after the Bonferroni correction (P <0.00022). Suggestive associations were found for centroid size, a proxy for dentoalveolar size variation with 4p16.1 and SNAI1. CONCLUSIONS Specific genetic pathways associated with 3-dimensional dentoalveolar phenotypic variation in malocclusions were identified.
Collapse
Affiliation(s)
| | - Steven F Miller
- Department of Anatomy, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, Ill; Department of Dental Medicine, College of Dental Medicine-Illinois, Midwestern University, Downers Grove, Ill
| | - Clarissa S G da Fontoura
- The Iowa Institute for Oral and Craniofacial Research, College of Dentistry, University of Iowa, Iowa City, Iowa
| | - George L Wehby
- Department of Health Management and Policy, College of Public Health, University of Iowa, Iowa City, Iowa
| | - Brad A Amendt
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Nathan E Holton
- Department of Orthodontics, College of Dentistry, University of Iowa, Iowa City, Iowa
| | - Veeratrishul Allareddy
- Department of Oral Pathology, Radiology and Medicine, College of Dentistry, University of Iowa, Iowa City, Iowa
| | - Thomas E Southard
- Department of Orthodontics, College of Dentistry, University of Iowa, Iowa City, Iowa
| | - Lina M Moreno Uribe
- The Iowa Institute for Oral and Craniofacial Research, College of Dentistry, University of Iowa, Iowa City, Iowa; Department of Orthodontics, College of Dentistry, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
21
|
Haillot E, Molina MD, Lapraz F, Lepage T. The Maternal Maverick/GDF15-like TGF-β Ligand Panda Directs Dorsal-Ventral Axis Formation by Restricting Nodal Expression in the Sea Urchin Embryo. PLoS Biol 2015; 13:e1002247. [PMID: 26352141 PMCID: PMC4564238 DOI: 10.1371/journal.pbio.1002247] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 08/05/2015] [Indexed: 01/26/2023] Open
Abstract
Specification of the dorsal-ventral axis in the highly regulative sea urchin embryo critically relies on the zygotic expression of nodal, but whether maternal factors provide the initial spatial cue to orient this axis is not known. Although redox gradients have been proposed to entrain the dorsal-ventral axis by acting upstream of nodal, manipulating the activity of redox gradients only has modest consequences, suggesting that other factors are responsible for orienting nodal expression and defining the dorsal-ventral axis. Here we uncover the function of Panda, a maternally provided transforming growth factor beta (TGF-β) ligand that requires the activin receptor-like kinases (Alk) Alk3/6 and Alk1/2 receptors to break the radial symmetry of the embryo and orient the dorsal-ventral axis by restricting nodal expression. We found that the double inhibition of the bone morphogenetic protein (BMP) type I receptors Alk3/6 and Alk1/2 causes a phenotype dramatically more severe than the BMP2/4 loss-of-function phenotype, leading to extreme ventralization of the embryo through massive ectopic expression of nodal, suggesting that an unidentified signal acting through BMP type I receptors cooperates with BMP2/4 to restrict nodal expression. We identified this ligand as the product of maternal Panda mRNA. Double inactivation of panda and bmp2/4 led to extreme ventralization, mimicking the phenotype caused by inactivation of the two BMP receptors. Inhibition of maternal panda mRNA translation disrupted the early spatial restriction of nodal, leading to persistent massive ectopic expression of nodal on the dorsal side despite the presence of Lefty. Phylogenetic analysis indicates that Panda is not a prototypical BMP ligand but a member of a subfamily of TGF-β distantly related to Inhibins, Lefty, and TGF-β that includes Maverick from Drosophila and GDF15 from vertebrates. Indeed, overexpression of Panda does not appear to directly or strongly activate phosphoSmad1/5/8 signaling, suggesting that although this TGF-β may require Alk1/2 and/or Alk3/6 to antagonize nodal expression, it may do so by sequestering a factor essential for Nodal signaling, by activating a non-Smad pathway downstream of the type I receptors, or by activating extremely low levels of pSmad1/5/8. We provide evidence that, although panda mRNA is broadly distributed in the early embryo, local expression of panda mRNA efficiently orients the dorsal-ventral axis and that Panda activity is required locally in the early embryo to specify this axis. Taken together, these findings demonstrate that maternal panda mRNA is both necessary and sufficient to orient the dorsal-ventral axis. These results therefore provide evidence that in the highly regulative sea urchin embryo, the activity of spatially restricted maternal factors regulates patterning along the dorsal-ventral axis.
Collapse
Affiliation(s)
- Emmanuel Haillot
- Institut de Biologie Valrose, iBV, UMR 7277 CNRS, Inserm U1091, UNS, University of Nice Sophia Antipolis, Nice, France
| | - Maria Dolores Molina
- Institut de Biologie Valrose, iBV, UMR 7277 CNRS, Inserm U1091, UNS, University of Nice Sophia Antipolis, Nice, France
| | - François Lapraz
- Institut de Biologie Valrose, iBV, UMR 7277 CNRS, Inserm U1091, UNS, University of Nice Sophia Antipolis, Nice, France
| | - Thierry Lepage
- Institut de Biologie Valrose, iBV, UMR 7277 CNRS, Inserm U1091, UNS, University of Nice Sophia Antipolis, Nice, France
| |
Collapse
|
22
|
TGFβ signaling in establishing left–right asymmetry. Semin Cell Dev Biol 2014; 32:80-4. [DOI: 10.1016/j.semcdb.2014.03.029] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 03/26/2014] [Indexed: 11/19/2022]
|
23
|
Nissimov JN, Das Chaudhuri AB. Hair curvature: a natural dialectic and review. Biol Rev Camb Philos Soc 2014; 89:723-66. [PMID: 24617997 DOI: 10.1111/brv.12081] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 12/18/2013] [Accepted: 01/01/2014] [Indexed: 12/19/2022]
Abstract
Although hair forms (straight, curly, wavy, etc.) are present in apparently infinite variations, each fibre can be reduced to a finite sequence of tandem segments of just three types: straight, bent/curly, or twisted. Hair forms can thus be regarded as resulting from genetic pathways that induce, reverse or modulate these basic curvature modes. However, physical interconversions between twists and curls demonstrate that strict one-to-one correspondences between them and their genetic causes do not exist. Current hair-curvature theories do not distinguish between bending and twisting mechanisms. We here introduce a multiple papillary centres (MPC) model which is particularly suitable to explain twisting. The model combines previously known features of hair cross-sectional morphology with partially/completely separated dermal papillae within single follicles, and requires such papillae to induce differential growth rates of hair cortical material in their immediate neighbourhoods. The MPC model can further help to explain other, poorly understood, aspects of hair growth and morphology. Separate bending and twisting mechanisms would be preferentially affected at the major or minor ellipsoidal sides of fibres, respectively, and together they exhaust the possibilities for influencing hair-form phenotypes. As such they suggest dialectic for hair-curvature development. We define a natural-dialectic (ND) which could take advantage of speculative aspects of dialectic, but would verify its input data and results by experimental methods. We use this as a top-down approach to first define routes by which hair bending or twisting may be brought about and then review evidence in support of such routes. In particular we consider the wingless (Wnt) and mammalian target of rapamycin (mTOR) pathways as paradigm pathways for molecular hair bending and twisting mechanisms, respectively. In addition to the Wnt canonical pathway, the Wnt/Ca(2+) and planar cell polarity (PCP) pathways, and others, can explain many alternatives and specific variations of hair bending phenotypes. Mechanisms for hair papilla budding or its division by bisection or fission can explain MPC formation. Epithelial-to-mesenchymal (EMT) and mesenchymal-to-epithelial (MET) transitions, acting in collaboration with epithelial-mesenchymal communications are also considered as mechanisms affecting hair growth and its bending and twisting. These may be treated as sub-mechanisms of an overall development from neural-crest stem cell (NCSC) lineages to differentiated hair follicle (HF) cell types, thus providing a unified framework for hair growth and development.
Collapse
|
24
|
Strizzi L, Postovit LM, Margaryan NV, Lipavsky A, Gadiot J, Blank C, Seftor RE, Seftor EA, Hendrix MJ. Nodal as a biomarker for melanoma progression and a new therapeutic target for clinical intervention. ACTA ACUST UNITED AC 2014; 4:67-78. [PMID: 19885369 DOI: 10.1586/17469872.4.1.67] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nodal, an embryonic morphogen belonging to the TGF-β superfamily, is an important regulator of embryonic stem cell fate. We have recently demonstrated that Nodal is expressed significantly in aggressive melanoma. Surprisingly, expression of the Nodal coreceptor, Cripto-1, was detected in only a small fraction of the melanoma tumor cell population, indicating a primary role for Cripto-1-independent signaling of Nodal in melanoma. In this review, we discuss how regulatory factors present in an embryonic environment, such as Lefty, can downregulate Nodal expression and inhibit tumorigenicity and plasticity of melanoma cells. Our translational studies show that antibodies against Nodal are capable of repressing melanoma vasculogenic mimicry and of inducing apoptosis in melanoma tumors in an in vivo lung-colonization assay. Our previous work and ongoing studies suggest that Nodal may represent a novel diagnostic marker and therapeutic target in melanoma.
Collapse
Affiliation(s)
- Luigi Strizzi
- Children's Memorial Research Center, 2300 Children's Plaza, Box 222, Chicago, IL 60614, USA Tel.: +1 773 755 6327
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Komatsu Y, Mishina Y. Establishment of left-right asymmetry in vertebrate development: the node in mouse embryos. Cell Mol Life Sci 2013; 70:4659-66. [PMID: 23771646 DOI: 10.1007/s00018-013-1399-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 05/30/2013] [Accepted: 05/31/2013] [Indexed: 01/20/2023]
Abstract
Establishment of vertebrate left-right asymmetry is a critical process for normal embryonic development. After the discovery of genes expressed asymmetrically along the left-right axis in chick embryos in the mid 1990s, the molecular mechanisms responsible for left-right patterning in vertebrate embryos have been studied extensively. In this review article, we discuss the mechanisms by which the initial symmetry along the left-right axis is broken in the mouse embryo. We focus on the role of primary cilia and molecular mechanisms of ciliogenesis at the node when symmetry is broken and left-right asymmetry is established. The node is considered a signaling center for early mouse embryonic development, and the results we review here have led to a better understanding of how the node functions and establishes left-right asymmetry.
Collapse
Affiliation(s)
- Yoshihiro Komatsu
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, 1011 N. University Ave., Ann Arbor, MI, 48109, USA
| | | |
Collapse
|
26
|
Development of the mammalian axial skeleton requires signaling through the Gα(i) subfamily of heterotrimeric G proteins. Proc Natl Acad Sci U S A 2012; 109:21366-71. [PMID: 23236180 DOI: 10.1073/pnas.1219810110] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
129/SvEv mice with a loss-of-function mutation in the heterotrimeric G protein α-subunit gene Gnai3 have fusions of ribs and lumbar vertebrae, indicating a requirement for Gα(i) (the "inhibitory" class of α-subunits) in somite derivatives. Mice with mutations of Gnai1 or Gnai2 have neither defect, but loss of both Gnai3 and one of the other two genes increases the number and severity of rib fusions without affecting the lumbar fusions. No myotome defects are observed in Gnai3/Gnai1 double-mutant embryos, and crosses with a conditional allele of Gnai2 indicate that Gα(i) is specifically required in cartilage precursors. Penetrance and expressivity of the rib fusion phenotype is altered in mice with a mixed C57BL/6 × 129/SvEv genetic background. These phenotypes reveal a previously unknown role for G protein-coupled signaling pathways in development of the axial skeleton.
Collapse
|
27
|
Katsu K, Tatsumi N, Niki D, Yamamura KI, Yokouchi Y. Multi-modal effects of BMP signaling on Nodal expression in the lateral plate mesoderm during left-right axis formation in the chick embryo. Dev Biol 2012. [PMID: 23206893 DOI: 10.1016/j.ydbio.2012.11.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During development of left-right asymmetry in the vertebrate embryo, Nodal plays a central role for determination of left-handedness. Bone morphogenetic protein (BMP) signaling has an important role for regulation of Nodal expression, although there is controversy over whether BMP signaling has a positive or negative effect on Nodal expression in the chick embryo. As BMP is a morphogen, we speculated that different concentrations might induce different responses in the cells of the lateral plate mesoderm (LPM). To test this hypothesis, we analyzed the effects of various concentrations of BMP4 and NOGGIN on Nodal expression in the LPM. We found that the effect on Nodal expression varied in a complex fashion with the concentration of BMP. In agreement with previous reports, we found that a high level of BMP signaling induced Nodal expression in the LPM, whereas a low level inhibited expression. However, a high intermediate level of BMP signaling was found to suppress Nodal expression in the left LPM, whereas a low intermediate level induced Nodal expression in the right LPM. Thus, the high and the low intermediate levels of BMP signaling up-regulated Nodal expression, but the high intermediate and low levels of BMP signaling down-regulated Nodal expression. Next, we sought to identify the mechanisms of this complex regulation of Nodal expression by BMP signaling. At the low intermediate level of BMP signaling, regulation depended on a NODAL positive-feedback loop suggesting the possibility of crosstalk between BMP and NODAL signaling. Overexpression of a constitutively active BMP receptor, a constitutively active ACTIVIN/NODAL receptor and SMAD4 indicated that SMAD1 and SMAD2 competed for binding to SMAD4 in the cells of the LPM. Nodal regulation by the high and low levels of BMP signaling was dependent on Cfc up-regulation or down-regulation, respectively. We propose a model for the variable effects of BMP signaling on Nodal expression in which different levels of BMP signaling regulate Nodal expression by a balance between BMP-pSMAD1/4 signaling and NODAL-pSMAD2/4 signaling.
Collapse
Affiliation(s)
- Kenjiro Katsu
- Division of Pattern Formation, Department of Organogenesis, Institute of Molecular Embryology and Genetics, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | | | | | | | | |
Collapse
|
28
|
Pereira PNG, Dobreva MP, Maas E, Cornelis FM, Moya IM, Umans L, Verfaillie CM, Camus A, de Sousa Lopes SMC, Huylebroeck D, Zwijsen A. Antagonism of Nodal signaling by BMP/Smad5 prevents ectopic primitive streak formation in the mouse amnion. Development 2012; 139:3343-54. [PMID: 22912414 DOI: 10.1242/dev.075465] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The strength and spatiotemporal activity of Nodal signaling is tightly controlled in early implantation mouse embryos, including by autoregulation and feedback loops, and involves secreted and intracellular antagonists. These control mechanisms, which are established at the extra-embryonic/embryonic interfaces, are essential for anterior-posterior patterning of the epiblast and correct positioning of the primitive streak. Formation of an ectopic primitive streak, or streak expansion, has previously been reported in mutants lacking antagonists that target Nodal signaling. Here, we demonstrate that loss-of-function of a major bone morphogenetic protein (BMP) effector, Smad5, results in formation of an ectopic primitive streak-like structure in mutant amnion accompanied by ectopic Nodal expression. This suggests that BMP/Smad5 signaling contributes to negative regulation of Nodal. In cultured cells, we find that BMP-activated Smad5 antagonizes Nodal signaling by interfering with the Nodal-Smad2/4-Foxh1 autoregulatory pathway through the formation of an unusual BMP4-induced Smad complex containing Smad2 and Smad5. Quantitative expression analysis supports that ectopic Nodal expression in the Smad5 mutant amnion is induced by the Nodal autoregulatory loop and a slow positive-feedback loop. The latter involves BMP4 signaling and also induction of ectopic Wnt3. Ectopic activation of these Nodal feedback loops in the Smad5 mutant amnion results in the eventual formation of an ectopic primitive streak-like structure. We conclude that antagonism of Nodal signaling by BMP/Smad5 signaling prevents primitive streak formation in the amnion of normal mouse embryos.
Collapse
Affiliation(s)
- Paulo N G Pereira
- Laboratory of Developmental Signaling of the VIB11 Center for the Biology of Disease, VIB, and Center for Human Genetics, KU Leuven, B-3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Nakamura T, Hamada H. Left-right patterning: conserved and divergent mechanisms. Development 2012; 139:3257-62. [PMID: 22912409 DOI: 10.1242/dev.061606] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The left-right (LR) asymmetry of visceral organs is fundamental to their function and position within the body. Over the past decade or so, the molecular mechanisms underlying the establishment of such LR asymmetry have been revealed in many vertebrate and invertebrate model organisms. These studies have identified a gene network that contributes to this process and is highly conserved from sea urchin to mouse. By contrast, some specific steps of the process, such as the symmetry-breaking event and situs-specific organogenesis, appear to have diverged during evolution. Here, we summarize the common and divergent mechanisms by which LR asymmetry is established in vertebrates.
Collapse
Affiliation(s)
- Tetsuya Nakamura
- Developmental Genetics Group, Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan.
| | | |
Collapse
|
30
|
Luo YJ, Su YH. Opposing nodal and BMP signals regulate left-right asymmetry in the sea urchin larva. PLoS Biol 2012; 10:e1001402. [PMID: 23055827 PMCID: PMC3467216 DOI: 10.1371/journal.pbio.1001402] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 08/28/2012] [Indexed: 12/28/2022] Open
Abstract
Nodal and BMP signals are important for establishing left-right (LR) asymmetry in vertebrates. In sea urchins, Nodal signaling prevents the formation of the rudiment on the right side. However, the opposing pathway to Nodal signaling during LR axis establishment is not clear. Here, we revealed that BMP signaling is activated in the left coelomic pouch, specifically in the veg2 lineage, but not in the small micromeres. By perturbing BMP activities, we demonstrated that BMP signaling is required for activating the expression of the left-sided genes and the formation of the left-sided structures. On the other hand, Nodal signals on the right side inhibit BMP signaling and control LR asymmetric separation and apoptosis of the small micromeres. Our findings show that BMP signaling is the positive signal for left-sided development in sea urchins, suggesting that the opposing roles of Nodal and BMP signals in establishing LR asymmetry are conserved in deuterostomes.
Collapse
Affiliation(s)
| | - Yi-Hsien Su
- Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei, Taiwan
| |
Collapse
|
31
|
Ao A, Hao J, Hopkins CR, Hong CC. DMH1, a novel BMP small molecule inhibitor, increases cardiomyocyte progenitors and promotes cardiac differentiation in mouse embryonic stem cells. PLoS One 2012; 7:e41627. [PMID: 22848549 PMCID: PMC3407188 DOI: 10.1371/journal.pone.0041627] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 06/22/2012] [Indexed: 01/24/2023] Open
Abstract
The possibility of using cell-based therapeutics to treat cardiac failure has generated significant interest since the initial introduction of stem cell-based technologies. However, the methods to quickly and robustly direct stem cell differentiation towards cardiac cell types have been limited by a reliance on recombinant growth factors to provide necessary biological cues. We report here the use of dorsomorphin homologue 1 (DMH1), a second-generation small molecule BMP inhibitor based on dorsomorphin, to efficiently induce beating cardiomyocyte formation in mouse embryonic stem cells (ESCs) and to specifically upregulate canonical transcriptional markers associated with cardiac development. DMH1 differs significantly from its predecessor by its ability to enrich for pro-cardiac progenitor cells that respond to late-stage Wnt inhibition using XAV939 and produce secondary beating cardiomyocytes. Our study demonstrates the utility of small molecules to complement existing in vitro cardiac differentiation protocols and highlights the role of transient BMP inhibition in cardiomyogenesis.
Collapse
Affiliation(s)
- Ada Ao
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- * E-mail: (AA); (CCH)
| | - Jijun Hao
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Corey R. Hopkins
- Vanderbilt Institute of Chemical Biology, Department of Pharmacology, Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Charles C. Hong
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Vanderbilt Institute of Chemical Biology, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, United States of America
- * E-mail: (AA); (CCH)
| |
Collapse
|
32
|
Robinson JF, Verhoef A, Piersma AH. Transcriptomic analysis of neurulation and early organogenesis in rat embryos: an in vivo and ex vivo comparison. Toxicol Sci 2012; 126:255-66. [PMID: 22262562 DOI: 10.1093/toxsci/kfr343] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cultured embryos mimic the morphological developmental progression of embryos (in vivo) undergoing neurulation and early organogenesis. Using available genomics technologies, comparative molecular-based assessments between cultured embryos and in vivo models may further clarify commonalities and dissimilarities, which contribute to differences between systems. Therefore, in this study, using a transcriptomic approach, we compared cultured whole rat embryos and embryos in vivo at comparable time points in development (gestational day (GD) 10 + 2-48 h, GD 0 = copulatory plug) to assess for commonalities and differences in gene expression in relation to morphology. We reveal strong parallels in time-dependent expression of genes in terms of magnitude, directionality, and functionality between whole embryo culture (WEC) and in vivo (rat). Genes changing in expression over time resemble previously hypothesized mechanisms underlying early development in mammalian systems. Furthermore, at the gene and functional level, we identify genes, which differ in expression between models, including genes related to development, oxygen transport, and metabolism. In summary, our results support the use of WEC for toxicological studies aimed at representing in vivo development during this time window at the molecular level. Additionally, we indicate genes, which differ in expression between models, providing possible insights for improvement of culture conditions.
Collapse
Affiliation(s)
- Joshua F Robinson
- Laboratory for Health Protection Research, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | | | | |
Collapse
|
33
|
Katsu K, Tokumori D, Tatsumi N, Suzuki A, Yokouchi Y. BMP inhibition by DAN in Hensen's node is a critical step for the establishment of left-right asymmetry in the chick embryo. Dev Biol 2011; 363:15-26. [PMID: 22202776 DOI: 10.1016/j.ydbio.2011.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 11/28/2022]
Abstract
During left-right (L-R) axis formation, Nodal is expressed in the node and has a central role in the transfer of L-R information in the vertebrate embryo. Bone morphogenetic protein (BMP) signaling also has an important role for maintenance of gene expression around the node. Several members of the Cerberus/Dan family act on L-R patterning by regulating activity of the transforming growth factor-β (TGF-β) family. We demonstrate here that chicken Dan plays a critical role in L-R axis formation. Chicken Dan is expressed in the left side of the node shortly after left-handed Shh expression and before the appearance of asymmetrically expressed genes in the lateral plate mesoderm (LPM). In vitro experiments revealed that DAN inhibited BMP signaling but not NODAL signaling. SHH had a positive regulatory effect on Dan expression while BMP4 had a negative effect. Using overexpression and RNA interference-mediated knockdown strategies, we demonstrate that Dan is indispensable for Nodal expression in the LPM and for Lefty-1 expression in the notochord. In the perinodal region, expression of Dan and Nodal was independent of each other. Nodal up-regulation by DAN required NODAL signaling, suggesting that DAN might act synergistically with NODAL. Our data indicate that Dan plays an essential role in the establishment of the L-R axis by inhibiting BMP signaling around the node.
Collapse
Affiliation(s)
- Kenjiro Katsu
- Division of Pattern Formation, Department of Organogenesis, Institute of Molecular Embryology and Genetics, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | | | | | | | | |
Collapse
|
34
|
Smith KA, Noël E, Thurlings I, Rehmann H, Chocron S, Bakkers J. Bmp and nodal independently regulate lefty1 expression to maintain unilateral nodal activity during left-right axis specification in zebrafish. PLoS Genet 2011; 7:e1002289. [PMID: 21980297 PMCID: PMC3183088 DOI: 10.1371/journal.pgen.1002289] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 07/30/2011] [Indexed: 11/21/2022] Open
Abstract
In vertebrates, left-right (LR) axis specification is determined by a ciliated structure in the posterior region of the embryo. Fluid flow in this ciliated structure is responsible for the induction of unilateral left-sided Nodal activity in the lateral plate mesoderm, which in turn regulates organ laterality. Bmp signalling activity has been implied in repressing Nodal expression on the right side, however its mechanism of action has been controversial. In a forward genetic screen for mutations that affect LR patterning, we identified the zebrafish linkspoot (lin) mutant, characterized by cardiac laterality and mild dorsoventral patterning defects. Mapping of the lin mutation revealed an inactivating missense mutation in the Bmp receptor 1aa (bmpr1aa) gene. Embryos with a mutation in lin/bmpr1aa and a novel mutation in its paralogue, bmpr1ab, displayed a variety of dorsoventral and LR patterning defects with increasing severity corresponding with a decrease in bmpr1a dosage. In Bmpr1a-deficient embryos we observed bilateral expression of the Nodal-related gene, spaw, coupled with reduced expression of the Nodal-antagonist lefty1 in the midline. Using genetic models to induce or repress Bmp activity in combination with Nodal inhibition or activation, we found that Bmp and Nodal regulate lefty1 expression in the midline independently of each other. Furthermore, we observed that the regulation of lefty1 by Bmp signalling is required for its observed downregulation of Nodal activity in the LPM providing a novel explanation for this phenomenon. From these results we propose a two-step model in which Bmp regulates LR patterning. Prior to the onset of nodal flow and Nodal activation, Bmp is required to induce lefty1 expression in the midline. When nodal flow has been established and Nodal activity is apparent, both Nodal and Bmp independently are required for lefty1 expression to assure unilateral Nodal activation and correct LR patterning. Although vertebrates are bilaterally symmetric when observed from the outside, inside the body cavity the organs are positioned asymmetrically with respect to the left and right sides. Cases where all the organs are mirror imaged, known as situs inversus, are not associated with any medical defects. Severe medical problems occur however in infants with a partial organ reversal (situs ambigious or heterotaxia), which arises during embryonic development. Left-right asymmetry in the embryo is established by unilateral expression of Nodal, a member of the Tgf-ß superfamily of secreted growth factors, a role that has been conserved from human to snails. By performing a genetic screen in zebrafish for laterality mutants, we have identified the linkspoot mutant, which displayed partial defects in asymmetric left-right positioning of the internal organs. The gene disrupted in the linkspoot mutant encodes a receptor for bone morphogenetic proteins (Bmp), another member of the Tgf-ß superfamily of secreted growth factors. Further analysis of Bmp over-expression or knock-down models demonstrate that Bmp signalling is required for unilateral Nodal expression, through the initiation and maintenance of an embryonic midline barrier. Our results demonstrate a novel and important mechanism by which left-right asymmetry in the vertebrate embryo is established and regulated.
Collapse
Affiliation(s)
- Kelly A. Smith
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Emily Noël
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ingrid Thurlings
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Holger Rehmann
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sonja Chocron
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
35
|
Myers M, Tripurani SK, Middlebrook B, Economides AN, Canalis E, Pangas SA. Loss of gremlin delays primordial follicle assembly but does not affect female fertility in mice. Biol Reprod 2011; 85:1175-82. [PMID: 21832168 DOI: 10.1095/biolreprod.111.091728] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The transforming growth factor beta (TGFB) protein family is renowned for its diverse roles in developmental biology including reproduction. Gremlin is a member of the differential screening-selected gene aberrative in neuroblastoma (DAN)/cerberus family of bone morphogenetic protein (BMP) antagonists. Recent studies on gremlin focus on its involvement in embryonic skeletal, lung, and kidney development. To define the role of gremlin (Grem1) in female reproduction, we analyzed postnatal folliculogenesis using global and conditional knockout (cKO) mice for gremlin. Grem1(-/-) mice die within 48 h after birth, and ovaries collected from neonatal Grem1(-/-) mice demonstrated reduced oocyte numbers and delayed primordial follicle development. Transplanting Grem1(-/-) neonatal ovaries showed that folliculogenesis proceeded to large antral follicle stage, but Grem1(-/-) ovaries contained corpora lutea-like structures not found in control-transplanted ovaries. However, Grem1 cKO mice had comparable fertility to control mice. These data suggest that gremlin plays a previously uncharacterized role in the regulation of oocyte numbers and the timing of primordial follicle development, but either it is not required for later folliculogenesis or its loss is possibly compensated by other BMP antagonists.
Collapse
Affiliation(s)
- Michelle Myers
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | |
Collapse
|
36
|
Komatsu Y, Kaartinen V, Mishina Y. Cell cycle arrest in node cells governs ciliogenesis at the node to break left-right symmetry. Development 2011; 138:3915-20. [PMID: 21831921 DOI: 10.1242/dev.068833] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cilia at the node generate a leftward fluid flow that breaks left-right symmetry. However, the molecular mechanisms that regulate ciliogenesis at the node are largely unknown. Here, we show that the epiblast-specific deletion of the gene encoding the BMP type 1 receptor (Acvr1) compromised development of nodal cilia, which results in defects in leftward fluid flow and, thus, abnormalities in left-right patterning. Acvr1 deficiency in mouse embryonic fibroblasts (MEFs) resulted in severe defects in their quiescence-induced primary cilia. Although the induction of quiescence in wild-type MEFs leads to an increase in the level of the cyclin-dependent kinase inhibitor p27(Kip1) and to rapid p27(Kip1) phosphorylation on Ser(10), MEFs deficient in Acvr1 show a reduction in both p27(Kip1) protein levels and in p27(Kip1) Ser(10) phosphorylation. The observed defects in cilium development were rescued by the introduction of p27(Kip1) into Acvr1-deficient MEFs, implying that BMP signaling positively controls p27(Kip1) stability in the G0 phase via p27(Kip1) Ser(10) phosphorylation, which is a prerequisite for induction of primary cilia. Importantly, in control embryos, p27(Kip1) protein is clearly present and strongly phosphorylated on Ser(10) in cells on the quiescent ventral surface of the node. By contrast, the corresponding cells in the node of Acvr1 mutant embryos were proliferative and showed a dramatic attenuation in both p27(Kip1) protein levels and phosphorylation on Ser(10). Our data suggest that cell quiescence controlled by BMP signaling via ACVR1 is required for transient formation of nodal cilia, and provide insight into the fundamental question of how the node represents the mechanistic `node' that regulates the development of left-right symmetry in vertebrates.
Collapse
Affiliation(s)
- Yoshihiro Komatsu
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, 1011 North University Avenue, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
37
|
Winzi MK, Hyttel P, Dale JK, Serup P. Isolation and characterization of node/notochord-like cells from mouse embryonic stem cells. Stem Cells Dev 2011; 20:1817-27. [PMID: 21351873 DOI: 10.1089/scd.2011.0042] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The homeobox gene Noto is expressed in the node and its derivative the notochord. Here we use a targeted Noto-GFP reporter to isolate and characterize node/notochord-like cells derived from mouse embryonic stem cells. We find very few Noto-expressing cells after spontaneous differentiation. However, the number of Noto-expressing cells was increased when using Activin A to induce a Foxa2- and Brachyury-expressing progenitor population, whose further differentiation into Noto-expressing cells was improved by simultaneous inhibition of BMP, Wnt, and retinoic acid signaling. Noto-GFP(+) cells expressed the node/notochord markers Noto, Foxa2, Shh, Noggin, Chordin, Foxj1, and Brachyury; showed a vacuolarization characteristic of notochord cells; and can integrate into midline structures when grafted into Hensen's node of gastrulating chicken embryos. The ability to generate node/notochord-like cells in vitro will aid the biochemical characterization of these developmentally important structures.
Collapse
Affiliation(s)
- Maria K Winzi
- Department of Stem Cell Biology, Hagedorn Research Institute, Gentofte, Denmark
| | | | | | | |
Collapse
|
38
|
Pulina MV, Hou SY, Mittal A, Julich D, Whittaker CA, Holley SA, Hynes RO, Astrof S. Essential roles of fibronectin in the development of the left-right embryonic body plan. Dev Biol 2011; 354:208-20. [PMID: 21466802 DOI: 10.1016/j.ydbio.2011.03.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 03/14/2011] [Accepted: 03/28/2011] [Indexed: 10/18/2022]
Abstract
Studies in Xenopus laevis suggested that cell-extracellular matrix (ECM) interactions regulate the development of the left-right axis of asymmetry; however, the identities of ECM components and their receptors important for this process have remained unknown. We discovered that FN is required for the establishment of the asymmetric gene expression pattern in early mouse embryos by regulating morphogenesis of the node, while cellular fates of the nodal cells, canonical Wnt and Shh signaling within the node were not perturbed by the absence of FN. FN is also required for the expression of Lefty 1/2 and activation of SMADs 2 and 3 at the floor plate, while cell fate specification of the notochord and the floor plate, as well as signaling within and between these two embryonic organizing centers remained intact in FN-null mutants. Furthermore, our experiments indicate that a major cell surface receptor for FN, integrin α5β1, is also required for the development of the left-right asymmetry, and that this requirement is evolutionarily conserved in fish and mice. Taken together, our studies demonstrate the requisite role for a structural ECM protein and its integrin receptor in the development of the left-right axis of asymmetry in vertebrates.
Collapse
Affiliation(s)
- Maria V Pulina
- Weill Cornell Medical College, Department of Medicine, Division of Cardiology, New York, NY, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Stottmann RW, Klingensmith J. Bone morphogenetic protein signaling is required in the dorsal neural folds before neurulation for the induction of spinal neural crest cells and dorsal neurons. Dev Dyn 2011; 240:755-65. [PMID: 21394823 DOI: 10.1002/dvdy.22579] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2011] [Indexed: 11/06/2022] Open
Abstract
Bone Morphogenetic Protein (BMP) activity has been implicated as a key regulator of multiple aspects of dorsal neural tube development. BMP signaling in the dorsal-most neuroepithelial cells presumably plays a critical role. We use tissue-specific gene ablation to probe the roles of BMPR1A, the type 1 BMP receptor that is seemingly the best candidate to mediate the activities of BMPs on early dorsal neural development. We use two different Cre lines expressed in the dorsal neural folds, one prior to spinal neurulation and one shortly afterward, together with a Bmpr1a conditional null mutation. Our findings indicate that BMPR1A signaling in the dorsal neural folds is important for hindbrain neural tube closure, but suggest it is dispensable for spinal neurulation. Our results also demonstrate a requirement for BMP signaling in patterning of dorsal neural tube cell fate and in neural crest cell formation, and imply a critical period shortly before neural tube closure.
Collapse
Affiliation(s)
- Rolf W Stottmann
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | | |
Collapse
|
40
|
Aamar E, Dawid IB. Sox17 and chordin are required for formation of Kupffer's vesicle and left-right asymmetry determination in zebrafish. Dev Dyn 2010; 239:2980-8. [PMID: 20925124 PMCID: PMC3090657 DOI: 10.1002/dvdy.22431] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Kupffer's vesicle (KV), a ciliated fluid-filled sphere in the zebrafish embryo with a critical role in laterality determination, is derived from a group of superficial cells in the organizer region of the gastrula named the dorsal forerunner cells (DFC). We have examined the role of the expression of sox17 and chordin (chd) in the DFC in KV formation and laterality determination. Whereas sox17 was known to be expressed in DFC, its function in these cells was not studied before. Further, expression of chd in these cells has not been reported previously. Targeted knockdown of Sox17 and Chd in DFC led to aberrant Left-Right (L-R) asymmetry establishment, as visualized by the expression of southpaw and lefty, and heart and pancreas placement in the embryo. These defects correlated with the formation of small KVs with apparently diminished cilia, consistent with the known requirement for ciliary function in the laterality organ for the establishment of L-R asymmetry.
Collapse
Affiliation(s)
| | - Igor B. Dawid
- Program in Genomics of Development, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda MD, USA
| |
Collapse
|
41
|
Yang YP, Anderson RM, Klingensmith J. BMP antagonism protects Nodal signaling in the gastrula to promote the tissue interactions underlying mammalian forebrain and craniofacial patterning. Hum Mol Genet 2010; 19:3030-42. [PMID: 20508035 DOI: 10.1093/hmg/ddq208] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Holoprosencephaly (HPE) is the most common forebrain and craniofacial malformation syndrome in humans. The genetics of HPE suggest that it often stems from a synergistic interaction of mutations in independent loci. In mice, several combinations of mutations in Nodal signaling pathway components can give rise to HPE, but it is not clear whether modest deficits of Nodal signaling along with lesions in other pathways might also cause such defects. We find that HPE results from simultaneous reduction of Nodal signaling and an organizer BMP (bone morphogenetic protein) antagonist, either Chordin or Noggin. These defects result from reduced production of tissues that promote forebrain and craniofacial development. Nodal promotes the expression of genes in the anterior primitive streak that are important for the development of these tissues, whereas BMP inhibits their expression. Pharmacological and transgenic manipulation of these signaling pathways suggests that BMP and Nodal antagonize each other prior to intracellular signal transduction. Biochemical experiments in vitro indicate that secreted Bmp2 and Nodal can form extracellular complexes, potentially interfering with receptor activation. Our results reveal that the patterning of forebrain and medial craniofacial elements requires a fine balance between BMP and Nodal signaling during primitive streak development, and provide a potential mechanistic basis for a new multigenic model of HPE.
Collapse
Affiliation(s)
- Yu-Ping Yang
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710-3709, USA
| | | | | |
Collapse
|
42
|
Stevens J, Ermakov A, Braganca J, Hilton H, Underhill P, Bhattacharya S, Brown NA, Norris DP. Analysis of the asymmetrically expressed Ablim1 locus reveals existence of a lateral plate Nodal-independent left sided signal and an early, left-right independent role for nodal flow. BMC DEVELOPMENTAL BIOLOGY 2010; 10:54. [PMID: 20487527 PMCID: PMC2885315 DOI: 10.1186/1471-213x-10-54] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Accepted: 05/20/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND Vertebrates show clear asymmetry in left-right (L-R) patterning of their organs and associated vasculature. During mammalian development a cilia driven leftwards flow of liquid leads to the left-sided expression of Nodal, which in turn activates asymmetric expression of the transcription factor Pitx2. While Pitx2 asymmetry drives many aspects of asymmetric morphogenesis, it is clear from published data that additional asymmetrically expressed loci must exist. RESULTS A L-R expression screen identified the cytoskeletally-associated gene, actin binding lim protein 1 (Ablim1), as asymmetrically expressed in both the node and left lateral plate mesoderm (LPM). LPM expression closely mirrors that of Nodal. Significantly, Ablim1 LPM asymmetry was detected in the absence of detectable Nodal. In the node, Ablim1 was initially expressed symmetrically across the entire structure, resolving to give a peri-nodal ring at the headfold stage in a flow and Pkd2-dependent manner. The peri-nodal ring of Ablim1 expression became asymmetric by the mid-headfold stage, showing stronger right than left-sided expression. Node asymmetry became more apparent as development proceeded; expression retreated in an anticlockwise direction, disappearing first from the left anterior node. Indeed, at early somite stages Ablim1 shows a unique asymmetric expression pattern, in the left lateral plate and to the right side of the node. CONCLUSION Left LPM Ablim1 is expressed in the absence of detectable LPM Nodal, clearly revealing existence of a Pitx2 and Nodal-independent left-sided signal in mammals. At the node, a previously unrecognised action of early nodal flow and Pkd2 activity, within the pit of the node, influences gene expression in a symmetric manner. Subsequent Ablim1 expression in the peri-nodal ring reveals a very early indication of L-R asymmetry. Ablim1 expression analysis at the node acts as an indicator of nodal flow. Together these results make Ablim1 a candidate for controlling aspects of L-R identity and patterning.
Collapse
Affiliation(s)
- Jonathan Stevens
- MRC Mammalian Genetics Unit, MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, UK
| | - Alexander Ermakov
- MRC Mammalian Genetics Unit, MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, UK
- Centre for Regenerative Medicine Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Jose Braganca
- Dept of Cardiovascular Medicine, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive Headington Oxford OX3 7BN, UK
- José Bragança Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
- IBB-Institute for Biotechnology and Bioengineering, Centro de Biomedicina Molecular e Estrutural, UAlg. Portugal
| | - Helen Hilton
- MRC Mammalian Genetics Unit, MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, UK
| | - Peter Underhill
- MRC Mammalian Genetics Unit, MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, UK
| | - Shoumo Bhattacharya
- Dept of Cardiovascular Medicine, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive Headington Oxford OX3 7BN, UK
| | - Nigel A Brown
- Division of Basic Medical Sciences, St George's University of London, Cranmer Terrace, London SW17 0RE, UK
| | - Dominic P Norris
- MRC Mammalian Genetics Unit, MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, UK
| |
Collapse
|
43
|
Dean DB, Watson JT, Jin W, Peters C, Enders JT, Chen A, Moed BR, Zhang Z. Distinct functionalities of bone morphogenetic protein antagonists during fracture healing in mice. J Anat 2010; 216:625-30. [PMID: 20298438 PMCID: PMC2871998 DOI: 10.1111/j.1469-7580.2010.01214.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2010] [Indexed: 11/29/2022] Open
Abstract
The bone morphogenetic protein (BMP) family of growth factors plays critical roles in bone formation. BMPs are regulated at multiple levels by various BMP antagonists. This study investigated how BMP antagonists are integrated into the cascade of events of bone formation during fracture healing. Forty mice underwent a controlled femur fracture; tissue samples at the fracture site were harvested at days 1, 3, 7, 14 and 21 after fracture, for quantification of the expression of BMPs and BMP antagonists. During fracture healing, BMP-2, -4 and -7 were up-regulated, but BMPR-1A and BMPR-2 showed reduced expression after day 14. Among BMP antagonists, the expressions of PRDC, SOST, Smad7, GREM1 and CERBERUS were generally down-regulated during fracture healing. In contrast, Noggin was significantly up-regulated in the first week after fracture; 7 days after fracture, other BMP antagonists, including DAN, CHRD, Smad6 and BAMBI, also showed significantly increased expression. In conclusion, this study indicates that BMP antagonists can be divided into two functional groups in relation to fracture healing: (1) those whose suppression may be essential for the initiation of osteogenesis; (2) those that are upregulated and may function in the remodeling of newly formed bone.
Collapse
Affiliation(s)
- Daniel B Dean
- Department of Orthopaedic Surgery, Saint Louis UniversitySt. Louis, MO, USA
| | - John T Watson
- Department of Orthopaedic Surgery, Saint Louis UniversitySt. Louis, MO, USA
| | - Wu Jin
- Department of Orthopaedic Surgery, Saint Louis UniversitySt. Louis, MO, USA
| | - Charlie Peters
- Anatomical Science Program, Saint Louis UniversitySt. Louis, MO, USA
| | - J T Enders
- Anatomical Science Program, Saint Louis UniversitySt. Louis, MO, USA
| | - Andrew Chen
- School of Medicine, Saint Louis UniversitySt. Louis, MO, USA
| | - Berton R Moed
- Department of Orthopaedic Surgery, Saint Louis UniversitySt. Louis, MO, USA
| | - Zijun Zhang
- Department of Orthopaedic Surgery, Saint Louis UniversitySt. Louis, MO, USA
- Anatomical Science Program, Saint Louis UniversitySt. Louis, MO, USA
| |
Collapse
|
44
|
Chen CM, Norris D, Bhattacharya S. Transcriptional control of left-right patterning in cardiac development. Pediatr Cardiol 2010; 31:371-7. [PMID: 20054532 DOI: 10.1007/s00246-009-9610-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2009] [Accepted: 12/07/2009] [Indexed: 10/20/2022]
Abstract
The heart develops from a simple left-right (L-R) symmetrical tube. Through a complex process of looping and remodelling, it becomes a highly L-R asymmetrical organ with distinct asymmetries in both morphology and function. Abnormal cardiac L-R patterning can result in a spectrum of defects that include, dextrocardia (a malposition of the heart to the right), isomerism of the atria (both atria being morphologically right-sided or left-sided), abnormal ventricular topology (e.g. the morphological left ventricle being dextral to the morphological right ventricle) or mirror-image topology (associated with situs inversus). Intermediate forms include abnormalities such as situs ambiguus and heterotaxia. L-R patterning abnormalities are typically associated with cardiac malformations, and it has become clear that an isolated septal, outflow tract and aortic arch malformation may be the only presenting manifestation of an L-R patterning defect. In the last two decades, there have been seminal advances in our understanding of the mechanisms controlling L-R patterning, and how mutations in L-R patterning genes result in human cardiac malformation. In this review, we provide an overview of the transcriptional mechanisms that result in asymmetric gene activation in mammals, how they receive information from signalling pathways, and how this translates to abnormal cardiac development.
Collapse
Affiliation(s)
- Chiann-mun Chen
- Department of Cardiovascular Medicine & Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
45
|
Vervoort R, Ceulemans H, Van Aerschot L, D'Hooge R, David G. Genetic modification of the inner ear lateral semicircular canal phenotype of the Bmp4 haplo-insufficient mouse. Biochem Biophys Res Commun 2010; 394:780-5. [PMID: 20233579 DOI: 10.1016/j.bbrc.2010.03.069] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Accepted: 03/10/2010] [Indexed: 12/27/2022]
Abstract
In the mouse, development of the lateral semicircular canal of the inner ear is sensitive to Bmp4 heterozygosity. In the C57BL6 background 30% of the heterozygotes display circling behavior, 66% have a specific defect in the vestibular part of the inner ear, namely the constriction, interruption or absence of the lateral semicircular canal. Only mice having both ears affected display circling behavior. In the (C57BL6xCBA)N1 background, the penetrance of the canal phenotype is greatly reduced, and bilateral lateral canal defect is not sufficient to induce circling. We found association of the canal phenotype with the genotype of markers on chromosome 14 and 4, co-localizing with Ecs and Eclb identified in the Ecl mouse with similar lateral canal defects. Candidate genes to contain the causal mutation are Bmp4 on chromosome 14, and Rere on chromosome 4.
Collapse
Affiliation(s)
- Raf Vervoort
- Laboratory of Glycobiology and Developmental Genetics, Flanders Institute for Biotechnology (VIB) and Department of Human Genetics, Katholieke Universiteit Leuven, Herestraat 49 bus 602, B-3000 Leuven, Belgium.
| | | | | | | | | |
Collapse
|
46
|
Tsiairis CD, McMahon AP. An Hh-dependent pathway in lateral plate mesoderm enables the generation of left/right asymmetry. Curr Biol 2009; 19:1912-7. [PMID: 19879143 DOI: 10.1016/j.cub.2009.09.057] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 09/10/2009] [Accepted: 09/10/2009] [Indexed: 12/01/2022]
Abstract
Breaking bilateral symmetry is critical for vertebrate morphogenesis. In the mouse, directional looping of the heart and rotation of the embryo, the first overt evidence of left/right asymmetry (L/R), are observed at early somite stages ( approximately E8.5) [1, 2]. Activation of a Nodal-Pitx2 regulatory pathway specifically within the left lateral plate mesoderm (LPM) is critical for these events [3-10]. Asymmetric expression of Nodal is thought to be triggered by left-oriented, cilia-generated flow within the ventral, midline node [11, 12]. Genetic removal of Hedgehog (Hh) signaling in the mouse demonstrates a requirement for Hedgehog signals in the symmetry-breaking process [13], and analysis of node trafficking has suggested a mechanism of directional transport in the node that might relate to symmetry breaking in the LPM [14]. Here we provide evidence that Hedgehog signaling in the node is not essential for breaking bilateral symmetry. In contrast, direct Hh signaling in the LPM is critical. Evidence is presented that Sonic and Indian hedgehog signals act together, through a Foxf1/Bmp4 pathway, to enable the initiation and propagation of Nodal signaling within the LPM, regulating the competence of that tissue to respond to the Nodal pathway.
Collapse
Affiliation(s)
- Charisios D Tsiairis
- Department of Molecular and Cellular Biology and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | | |
Collapse
|
47
|
Wu MY, Hill CS. Tgf-beta superfamily signaling in embryonic development and homeostasis. Dev Cell 2009; 16:329-43. [PMID: 19289080 DOI: 10.1016/j.devcel.2009.02.012] [Citation(s) in RCA: 570] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
TGF-beta superfamily signaling pathways emerged with the evolution of multicellular animals, suggesting that these pathways contribute to the increased diversity and complexity required for the development and homeostasis of these organisms. In this review we begin by exploring some key developmental and disease processes requiring TGF-beta ligands to underscore the fundamental importance of these pathways before delving into the molecular mechanism of signal transduction, focusing on recent findings. Finally, we discuss how these ligands act as morphogens, how their activity and signaling range is regulated, and how they interact with other signaling pathways to achieve their specific and varied functional roles.
Collapse
Affiliation(s)
- Mary Y Wu
- Laboratory of Developmental Signalling, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3PX, UK
| | | |
Collapse
|
48
|
Furtado MB, Solloway MJ, Jones VJ, Costa MW, Biben C, Wolstein O, Preis JI, Sparrow DB, Saga Y, Dunwoodie SL, Robertson EJ, Tam PPL, Harvey RP. BMP/SMAD1 signaling sets a threshold for the left/right pathway in lateral plate mesoderm and limits availability of SMAD4. Genes Dev 2009; 22:3037-49. [PMID: 18981480 DOI: 10.1101/gad.1682108] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Bistability in developmental pathways refers to the generation of binary outputs from graded or noisy inputs. Signaling thresholds are critical for bistability. Specification of the left/right (LR) axis in vertebrate embryos involves bistable expression of transforming growth factor beta (TGFbeta) member NODAL in the left lateral plate mesoderm (LPM) controlled by feed-forward and feedback loops. Here we provide evidence that bone morphogenetic protein (BMP)/SMAD1 signaling sets a repressive threshold in the LPM essential for the integrity of LR signaling. Conditional deletion of Smad1 in the LPM led to precocious and bilateral pathway activation. NODAL expression from both the left and right sides of the node contributed to bilateral activation, indicating sensitivity of mutant LPM to noisy input from the LR system. In vitro, BMP signaling inhibited NODAL pathway activation and formation of its downstream SMAD2/4-FOXH1 transcriptional complex. Activity was restored by overexpression of SMAD4 and in embryos, elevated SMAD4 in the right LPM robustly activated LR gene expression, an effect reversed by superactivated BMP signaling. We conclude that BMP/SMAD1 signaling sets a bilateral, repressive threshold for NODAL-dependent Nodal activation in LPM, limiting availability of SMAD4. This repressive threshold is essential for bistable output of the LR system.
Collapse
Affiliation(s)
- Milena B Furtado
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Differential requirement for BMP signaling in atrial and ventricular lineages establishes cardiac chamber proportionality. Dev Biol 2009; 328:472-82. [PMID: 19232521 DOI: 10.1016/j.ydbio.2009.02.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 02/04/2009] [Accepted: 02/06/2009] [Indexed: 11/23/2022]
Abstract
The function of an organ relies upon the proper relative proportions of its individual operational components. For example, effective embryonic circulation requires the appropriate relative sizes of each of the distinct pumps created by the atrial and ventricular cardiac chambers. Although the differences between atrial and ventricular cardiomyocytes are well established, little is known about the mechanisms regulating production of proportional numbers of each cell type. We find that mutation of the zebrafish type I BMP receptor gene alk8 causes reduction of atrial size without affecting the ventricle. Loss of atrial tissue is evident in the lateral mesoderm prior to heart tube formation and results from the inhibition of BMP signaling during cardiac progenitor specification stages. Comparison of the effects of decreased and increased BMP signaling further demonstrates that atrial cardiomyocyte production correlates with levels of BMP signaling while ventricular cardiomyocyte production is less susceptible to manipulation of BMP signaling. Additionally, mosaic analysis provides evidence for a cell-autonomous requirement for BMP signaling during cardiomyocyte formation and chamber fate assignment. Together, our studies uncover a new role for BMP signaling in the regulation of chamber size, supporting a model in which differential reception of cardiac inductive signals establishes chamber proportion.
Collapse
|
50
|
Yu X, He F, Zhang T, Espinoza-Lewis RA, Lin L, Yang J, Chen Y. Cerberus functions as a BMP agonist to synergistically induce nodal expression during left-right axis determination in the chick embryo. Dev Dyn 2009; 237:3613-23. [PMID: 18985739 DOI: 10.1002/dvdy.21769] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Left-sided expression of Nodal in the lateral plate mesoderm (LPM) during early embryogenesis is a crucial step in establishing the left-right (L-R) axis in vertebrates. In the chick, it was suggested that chick Cerberus (cCer), a Cerberus/Dan family member, induces Nodal expression by antagonizing bone morphogenetic protein (BMP) activity in the left LPM. In contrast, it has also been shown that BMPs positively regulate Nodal expression in the left LPM in the chick embryo. Thus, it is still unclear how the bilaterally expressed BMPs induce Nodal expression only in the left LPM. In this study, we demonstrate that BMP signaling is necessary and sufficient for the induction of Nodal expression in the chick LPM where the type I BMP receptor-IB (BMPR-IB) likely mediates this induction. Tissue grafting experiments indicate the existence of a Nodal inductive factor in the left LPM rather than the presence of a Nodal inhibitory factor in the right LPM. We demonstrate that cCer functions as a BMP agonist instead of antagonist, being able to enhance BMP signaling in cell culture. This conclusion is further supported by the immunoprecipitation assays that provide convincing biochemical evidence for a direct interaction between cCer and BMP receptor. Because cCer is expressed restrictedly in the left LPM, BMPs and cCer appear to act synergistically to activate Nodal expression in the left LPM in the chick.
Collapse
Affiliation(s)
- Xueyan Yu
- Section of Oral Biology, The Ohio State University College of Dentistry, Columbus, Ohio 43210, USA.
| | | | | | | | | | | | | |
Collapse
|