Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Development ; 150(18)2023 09 15.
Article in English | MEDLINE | ID: mdl-37680191

ABSTRACT

During zebrafish heart formation, cardiac progenitor cells converge at the embryonic midline where they form the cardiac cone. Subsequently, this structure transforms into a heart tube. Little is known about the molecular mechanisms that control these morphogenetic processes. Here, we use light-sheet microscopy and combine genetic, molecular biological and pharmacological tools to show that the paralogous genes wnt9a/b are required for the assembly of the nascent heart tube. In wnt9a/b double mutants, cardiomyocyte progenitor cells are delayed in their convergence towards the embryonic midline, the formation of the heart cone is impaired and the transformation into an elongated heart tube fails. The same cardiac phenotype occurs when both canonical and non-canonical Wnt signaling pathways are simultaneously blocked by pharmacological inhibition. This demonstrates that Wnt9a/b and canonical and non-canonical Wnt signaling regulate the migration of cardiomyocyte progenitor cells and control the formation of the cardiac tube. This can be partly attributed to their regulation of the timing of cardiac progenitor cell differentiation. Our study demonstrates how these morphogens activate a combination of downstream pathways to direct cardiac morphogenesis.


Subject(s)
Myocytes, Cardiac , Zebrafish , Animals , Zebrafish/genetics , Wnt Signaling Pathway/genetics , Cell Differentiation/genetics , Microscopy , Zebrafish Proteins/genetics , Wnt Proteins/genetics
2.
Mol Biol Evol ; 41(2)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38410843

ABSTRACT

In the African weakly electric fish genus Campylomormyrus, electric organ discharge signals are strikingly different in shape and duration among closely related species, contribute to prezygotic isolation, and may have triggered an adaptive radiation. We performed mRNA sequencing on electric organs and skeletal muscles (from which the electric organs derive) from 3 species with short (0.4 ms), medium (5 ms), and long (40 ms) electric organ discharges and 2 different cross-species hybrids. We identified 1,444 upregulated genes in electric organ shared by all 5 species/hybrid cohorts, rendering them candidate genes for electric organ-specific properties in Campylomormyrus. We further identified several candidate genes, including KCNJ2 and KLF5, and their upregulation may contribute to increased electric organ discharge duration. Hybrids between a short (Campylomormyrus compressirostris) and a long (Campylomormyrus rhynchophorus) discharging species exhibit electric organ discharges of intermediate duration and showed imbalanced expression of KCNJ2 alleles, pointing toward a cis-regulatory difference at this locus, relative to electric organ discharge duration. KLF5 is a transcription factor potentially balancing potassium channel gene expression, a crucial process for the formation of an electric organ discharge. Unraveling the genetic basis of the species-specific modulation of the electric organ discharge in Campylomormyrus is crucial for understanding the adaptive radiation of this emerging model taxon of ecological (perhaps even sympatric) speciation.


Subject(s)
Electric Fish , Animals , Electric Fish/genetics , Alleles , Electric Organ/metabolism , Up-Regulation , Potassium Channels/genetics
3.
J Cell Sci ; 134(1)2021 01 11.
Article in English | MEDLINE | ID: mdl-33323504

ABSTRACT

Steinberg's differential adhesion hypothesis suggests that adhesive mechanisms are important for sorting of cells and tissues during morphogenesis (Steinberg, 2007). During zebrafish vasculogenesis, endothelial cells sort into arterial and venous vessel beds but it is unknown whether this involves adhesive mechanisms. Claudins are tight junction proteins regulating the permeability of epithelial and endothelial tissue barriers. Previously, the roles of claudins during organ development have exclusively been related to their canonical functions in determining paracellular permeability. Here, we use atomic force microscopy to quantify claudin-5-dependent adhesion and find that this strongly contributes to the adhesive forces between arterial endothelial cells. Based on genetic manipulations, we reveal a non-canonical role of Claudin-5a during zebrafish vasculogenesis, which involves the regulation of adhesive forces between adjacent dorsal aortic endothelial cells. In vitro and in vivo studies demonstrate that loss of claudin-5 results in increased motility of dorsal aorta endothelial cells and in a failure of the dorsal aorta to lumenize. Our findings uncover a novel role of claudin-5 in limiting arterial endothelial cell motility, which goes beyond its traditional sealing function during embryonic development.


Subject(s)
Tight Junction Proteins , Tight Junctions , Animals , Claudin-4 , Claudin-5/genetics , Claudins , Endothelial Cells , Zebrafish , Zebrafish Proteins
4.
Development ; 147(16)2020 08 25.
Article in English | MEDLINE | ID: mdl-32843528

ABSTRACT

The Hippo-Yap pathway regulates multiple cellular processes in response to mechanical and other stimuli. In Drosophila, the polarity protein Lethal (2) giant larvae [L(2)gl], negatively regulates Hippo-mediated transcriptional output. However, in vertebrates, little is known about its homolog Llgl1. Here, we define a novel role for vertebrate Llgl1 in regulating Yap stability in cardiomyocytes, which impacts heart development. In contrast to the role of Drosophila L(2)gl, Llgl1 depletion in cultured rat cardiomyocytes decreased Yap protein levels and blunted target gene transcription without affecting Yap transcript abundance. Llgl1 depletion in zebrafish resulted in larger and dysmorphic cardiomyocytes, pericardial effusion, impaired blood flow and aberrant valvulogenesis. Cardiomyocyte Yap protein levels were decreased in llgl1 morphants, whereas Notch, which is regulated by hemodynamic forces and participates in valvulogenesis, was more broadly activated. Consistent with the role of Llgl1 in regulating Yap stability, cardiomyocyte-specific overexpression of Yap in Llgl1-depleted embryos ameliorated pericardial effusion and restored blood flow velocity. Altogether, our data reveal that vertebrate Llgl1 is crucial for Yap stability in cardiomyocytes and its absence impairs cardiac development.


Subject(s)
Cell Cycle Proteins/metabolism , Heart/embryology , Myocytes, Cardiac/metabolism , Trans-Activators/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cell Cycle Proteins/genetics , Protein Stability , Trans-Activators/genetics , YAP-Signaling Proteins , Zebrafish/genetics , Zebrafish Proteins/genetics
5.
Development ; 146(22)2019 11 14.
Article in English | MEDLINE | ID: mdl-31628109

ABSTRACT

Cardiac looping is an essential and highly conserved morphogenetic process that places the different regions of the developing vertebrate heart tube into proximity of their final topographical positions. High-resolution 4D live imaging of mosaically labelled cardiomyocytes reveals distinct cardiomyocyte behaviors that contribute to the deformation of the entire heart tube. Cardiomyocytes acquire a conical cell shape, which is most pronounced at the superior wall of the atrioventricular canal and contributes to S-shaped bending. Torsional deformation close to the outflow tract contributes to a torque-like winding of the entire heart tube between its two poles. Anisotropic growth of cardiomyocytes based on their positions reinforces S-shaping of the heart. During cardiac looping, bone morphogenetic protein pathway signaling is strongest at the future superior wall of the atrioventricular canal. Upon pharmacological or genetic inhibition of bone morphogenetic protein signaling, myocardial cells at the superior wall of the atrioventricular canal maintain cuboidal cell shapes and S-shaped bending is impaired. This description of cellular rearrangements and cardiac looping regulation may also be relevant for understanding the etiology of human congenital heart defects.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Gene Expression Regulation, Developmental , Heart/embryology , Myocytes, Cardiac/metabolism , Signal Transduction , Animals , Anisotropy , Embryo, Nonmammalian/metabolism , Embryonic Development , Green Fluorescent Proteins/metabolism , Microscopy, Confocal , Morphogenesis , Organogenesis , Torque , Transcription Factors/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway , Zebrafish/embryology , Zebrafish Proteins/metabolism
6.
Curr Opin Hematol ; 28(3): 198-207, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33714969

ABSTRACT

PURPOSE OF REVIEW: The zebrafish embryo has emerged as a powerful model organism to investigate the mechanisms by which biophysical forces regulate vascular and cardiac cell biology during development and disease. A versatile arsenal of methods and tools is available to manipulate and analyze biomechanical signaling. This review aims to provide an overview of the experimental strategies and tools that have been utilized to study biomechanical signaling in cardiovascular developmental processes and different vascular disease models in the zebrafish embryo. Within the scope of this review, we focus on work published during the last two years. RECENT FINDINGS: Genetic and pharmacological tools for the manipulation of cardiac function allow alterations of hemodynamic flow patterns in the zebrafish embryo and various types of transgenic lines are available to report endothelial cell responses to biophysical forces. These tools have not only revealed the impact of biophysical forces on cardiovascular development but also helped to establish more accurate models for cardiovascular diseases including cerebral cavernous malformations, hereditary hemorrhagic telangiectasias, arteriovenous malformations, and lymphangiopathies. SUMMARY: The zebrafish embryo is a valuable vertebrate model in which in-vivo manipulations of biophysical forces due to cardiac contractility and blood flow can be performed. These analyses give important insights into biomechanical signaling pathways that control endothelial and endocardial cell behaviors. The technical advances using this vertebrate model will advance our understanding of the impact of biophysical forces in cardiovascular pathologies.


Subject(s)
Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular System/embryology , Cardiovascular System/metabolism , Mechanotransduction, Cellular , Organogenesis , Signal Transduction , Zebrafish , Animals , Animals, Genetically Modified , Disease Susceptibility , Humans , Models, Animal
8.
Circ Res ; 125(10): e43-e54, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31495257

ABSTRACT

RATIONALE: Pathological biomechanical signaling induces vascular anomalies including cerebral cavernous malformations (CCM), which are caused by a clonal loss of CCM1/KRIT1 (Krev interaction trapped protein 1), CCM2/MGC4607, or CCM3/PDCD10. Why patients typically experience lesions only in lowly perfused venous capillaries of the cerebrovasculature is completely unknown. OBJECTIVE: In contrast, animal models with a complete loss of CCM proteins lack a functional heart and blood flow and exhibit vascular anomalies within major blood vessels as well. This finding raises the possibility that hemodynamics may play a role in the context of this vascular pathology. METHODS AND RESULTS: Here, we used a genetic approach to restore cardiac function and blood flow in a zebrafish model of CCM1. We find that blood flow prevents cardiovascular anomalies including a hyperplastic expansion within a large Ccm1-deficient vascular bed, the lateral dorsal aorta. CONCLUSIONS: This study identifies blood flow as an important physiological factor that is protective in the cause of this devastating vascular pathology.


Subject(s)
Blood Flow Velocity/physiology , Central Nervous System Neoplasms/diagnostic imaging , Disease Models, Animal , Hemangioma, Cavernous, Central Nervous System/diagnostic imaging , Animals , Animals, Genetically Modified , Central Nervous System Neoplasms/physiopathology , Cerebral Angiography/methods , Hemangioma, Cavernous, Central Nervous System/physiopathology , Zebrafish
9.
Stroke ; 51(4): 1272-1278, 2020 04.
Article in English | MEDLINE | ID: mdl-31992178

ABSTRACT

Background and Purpose- Cerebral cavernous malformations (CCMs) are vascular malformations of the brain that lead to cerebral hemorrhages. A pharmacological treatment is needed especially for patients with nonoperable deep-seated lesions. We and others obtained CCM mouse models that were useful for mechanistic studies and rapid trials testing the preventive effects of candidate drugs. The shortened lifespan of acute mouse models hampered evaluation of compounds that would not only prevent lesion appearance but also cure preexisting lesions. Indirubin-3'-monoxime previously demonstrated its efficacy to reverse the cardiac phenotype of ccm2m201 zebrafish mutants and to prevent lesion development in an acute CCM2 mouse model. In the present article, we developed and characterized a novel chronic CCM2 mouse model and evaluated the curative therapeutic effect of indirubin-3'-monoxime after CCM lesion development. Methods- The chronic mouse model was obtained by a postnatal induction of brain-endothelial-cell-specific ablation of the Ccm2 gene using the inducible Slco1c1-CreERT2 mouse line. Results- We obtained a fully penetrant novel CCM chronic mouse model without any obvious off-target phenotypes and compatible with long-term survival. By 3 months of age, CCM lesions ranging in size from small isolated lesions to multiple caverns developed throughout the brain. Lesion burden was quantified in animals from 1 week to 5 months of age. Clear signs of intracerebral hemorrhages were noticed in brain-endothelial-cell-specific ablation of the Ccm2 gene. In contrast with its preventive effect in the acute CCM2 mouse model, a 20 mg/kg indirubin-3'-monoxime treatment for 3 weeks in 3-month old animals neither had any beneficial effect on the lesion burden nor alleviated cerebral hemorrhages. Conclusions- The brain-endothelial-cell-specific ablation of the Ccm2 gene chronic model is a strongly improved disease model for the CCM community whose challenge today is to decipher which candidate drugs might have a curative effect on patients' preexisting lesions. Visual Overview- An online visual overview is available for this article.


Subject(s)
Brain/pathology , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Disease Models, Animal , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/pathology , Microfilament Proteins/genetics , Animals , Central Nervous System Neoplasms/metabolism , Hemangioma, Cavernous, Central Nervous System/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/deficiency
10.
J Cell Sci ; 131(15)2018 08 13.
Article in English | MEDLINE | ID: mdl-30030370

ABSTRACT

Endothelial integrity relies on a mechanical crosstalk between intercellular and cell-matrix interactions. This crosstalk is compromised in hemorrhagic vascular lesions of patients carrying loss-of-function mutations in cerebral cavernous malformation (CCM) genes. RhoA/ROCK-dependent cytoskeletal remodeling is central to the disease, as it causes unbalanced cell adhesion towards increased cell-extracellular matrix adhesions and destabilized cell-cell junctions. This study reveals that CCM proteins directly orchestrate ROCK1 and ROCK2 complementary roles on the mechanics of the endothelium. CCM proteins act as a scaffold, promoting ROCK2 interactions with VE-cadherin and limiting ROCK1 kinase activity. Loss of CCM1 (also known as KRIT1) produces excessive ROCK1-dependent actin stress fibers and destabilizes intercellular junctions. Silencing of ROCK1 but not ROCK2 restores the adhesive and mechanical homeostasis of CCM1 and CCM2-depleted endothelial monolayers, and rescues the cardiovascular defects of ccm1 mutant zebrafish embryos. Conversely, knocking down Rock2 but not Rock1 in wild-type zebrafish embryos generates defects reminiscent of the ccm1 mutant phenotypes. Our study uncovers the role of the CCM1-CCM2 complex in controlling ROCK1 and ROCK2 to preserve endothelial integrity and drive heart morphogenesis. Moreover, it solely identifies the ROCK1 isoform as a potential therapeutic target for the CCM disease.


Subject(s)
Carrier Proteins/metabolism , Endothelial Cells/metabolism , KRIT1 Protein/metabolism , rho-Associated Kinases/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Carrier Proteins/genetics , Cattle , Endothelial Cells/cytology , Flow Cytometry , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells , Humans , Immunoprecipitation , KRIT1 Protein/genetics , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish , rho-Associated Kinases/genetics
11.
Chemistry ; 26(65): 14838-14843, 2020 Nov 20.
Article in English | MEDLINE | ID: mdl-32501570

ABSTRACT

Oxidation of protein methionines to methionine-sulfoxides (MetOx) is associated with several age-related diseases. In healthy cells, MetOx is reduced to methionine by two families of conserved methionine sulfoxide reductase enzymes, MSRA and MSRB that specifically target the S- or R-diastereoisomers of methionine-sulfoxides, respectively. To directly interrogate MSRA and MSRB functions in cellular settings, we developed an NMR-based biosensor that we call CarMetOx to simultaneously measure both enzyme activities in single reaction setups. We demonstrate the suitability of our strategy to delineate MSR functions in complex biological environments, including cell lysates and live zebrafish embryos. Thereby, we establish differences in substrate specificities between prokaryotic and eukaryotic MSRs and introduce CarMetOx as a highly sensitive tool for studying therapeutic targets of oxidative stress-related human diseases and redox regulated signaling pathways.


Subject(s)
Biosensing Techniques , Humans , Methionine , Methionine Sulfoxide Reductases/metabolism , Oxidation-Reduction , Substrate Specificity
12.
Development ; 143(3): 373-86, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26839341

ABSTRACT

Endocardial cells are cardiac endothelial cells that line the interior of the heart tube. Historically, their contribution to cardiac development has mainly been considered from a morphological perspective. However, recent studies have begun to define novel instructive roles of the endocardium, as a sensor and signal transducer of biophysical forces induced by blood flow, and as an angiocrine signalling centre that is involved in myocardial cellular morphogenesis, regeneration and reprogramming. In this Review, we discuss how the endocardium develops, how endocardial-myocardial interactions influence the developing embryonic heart, and how the dysregulation of blood flow-responsive endocardial signalling can result in pathophysiological changes.


Subject(s)
Endocardium/embryology , Mechanotransduction, Cellular , Morphogenesis , Animals , Hemodynamics , Humans , Regeneration , Stem Cells/cytology
13.
Nucleic Acids Res ; 44(6): 2538-53, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26582913

ABSTRACT

DPF3 (BAF45c) is a member of the BAF chromatin remodeling complex. Two isoforms have been described, namely DPF3a and DPF3b. The latter binds to acetylated and methylated lysine residues of histones. Here, we elaborate on the role of DPF3a and describe a novel pathway of cardiac gene transcription leading to pathological cardiac hypertrophy. Upon hypertrophic stimuli, casein kinase 2 phosphorylates DPF3a at serine 348. This initiates the interaction of DPF3a with the transcriptional repressors HEY, followed by the release of HEY from the DNA. Moreover, BRG1 is bound by DPF3a, and is thus recruited to HEY genomic targets upon interaction of the two components. Consequently, the transcription of downstream targets such as NPPA and GATA4 is initiated and pathological cardiac hypertrophy is established. In human, DPF3a is significantly up-regulated in hypertrophic hearts of patients with hypertrophic cardiomyopathy or aortic stenosis. Taken together, we show that activation of DPF3a upon hypertrophic stimuli switches cardiac fetal gene expression from being silenced by HEY to being activated by BRG1. Thus, we present a novel pathway for pathological cardiac hypertrophy, whose inhibition is a long-term therapeutic goal for the treatment of the course of heart failure.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cardiomegaly/genetics , Chromatin Assembly and Disassembly , Chromatin/chemistry , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Differentiation , Chromatin/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Myoblasts/cytology , Myoblasts/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nuclear Proteins/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic
15.
Development ; 140(7): 1550-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23482490

ABSTRACT

Motile cilia perform crucial functions during embryonic development and throughout adult life. Development of organs containing motile cilia involves regulation of cilia formation (ciliogenesis) and formation of a luminal space (lumenogenesis) in which cilia generate fluid flows. Control of ciliogenesis and lumenogenesis is not yet fully understood, and it remains unclear whether these processes are coupled. In the zebrafish embryo, lethal giant larvae 2 (lgl2) is expressed prominently in ciliated organs. Lgl proteins are involved in establishing cell polarity and have been implicated in vesicle trafficking. Here, we identified a role for Lgl2 in development of ciliated epithelia in Kupffer's vesicle, which directs left-right asymmetry of the embryo; the otic vesicles, which give rise to the inner ear; and the pronephric ducts of the kidney. Using Kupffer's vesicle as a model ciliated organ, we found that depletion of Lgl2 disrupted lumen formation and reduced cilia number and length. Immunofluorescence and time-lapse imaging of Kupffer's vesicle morphogenesis in Lgl2-deficient embryos suggested cell adhesion defects and revealed loss of the adherens junction component E-cadherin at lateral membranes. Genetic interaction experiments indicate that Lgl2 interacts with Rab11a to regulate E-cadherin and mediate lumen formation that is uncoupled from cilia formation. These results uncover new roles and interactions for Lgl2 that are crucial for both lumenogenesis and ciliogenesis and indicate that these processes are genetically separable in zebrafish.


Subject(s)
Cilia/physiology , Kupffer Cells/physiology , Morphogenesis/genetics , Zebrafish Proteins/physiology , Zebrafish , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Polarity/genetics , Cilia/genetics , Cilia/metabolism , Embryo, Nonmammalian , Embryonic Development/genetics , Embryonic Development/physiology , Gene Expression Regulation, Developmental , Kupffer Cells/metabolism , Larva/genetics , Larva/metabolism , Morphogenesis/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
16.
Biochem Biophys Res Commun ; 468(4): 708-12, 2015 Dec 25.
Article in English | MEDLINE | ID: mdl-26551461

ABSTRACT

The cyclic pyrimidine nucleotides cCMP and cUMP occur in mammalian cell lines. Recently, cCMP was also identified in mouse organs. Due to technical difficulties, it has not been possible to detect cUMP in organs or tissues yet. Here, we have generated a temporal profile of the occurrence of nucleoside 3',5'-cyclic monophosphates during different developmental stages of embryogenesis and in different organs of the adult zebrafish Danio rerio. Cyclic nucleotides were quantified by high performance liquid chromatography quadrupole tandem mass spectrometry. The identity of cCMP and cUMP in the zebrafish was confirmed by high performance liquid chromatography quadrupole time-of-flight mass spectrometry. We show for the first time that cUMP can be detected during embryogenesis and in adult organs of this vertebrate model system.


Subject(s)
Cyclic CMP/metabolism , Embryonic Development/physiology , Nucleotides, Cyclic/metabolism , Uridine Monophosphate/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Aging/physiology , Animals , Gene Expression Regulation, Developmental/physiology , Organ Specificity/physiology , Tissue Distribution
17.
Development ; 138(10): 1935-45, 2011 May.
Article in English | MEDLINE | ID: mdl-21471154

ABSTRACT

Endodermal organogenesis requires a precise orchestration of cell fate specification and cell movements, collectively coordinating organ size and shape. In Caenorhabditis elegans, uncoordinated-53 (unc-53) encodes a neural guidance molecule that directs axonal growth. One of the vertebrate homologs of unc-53 is neuron navigator 3 (Nav3). Here, we identified a novel vertebrate neuron navigator 3 isoform in zebrafish, nav3a, and we provide genetic evidence in loss- and gain-of-function experiments showing its functional role in endodermal organogenesis during zebrafish embryogenesis. In zebrafish embryos, nav3a expression was initiated at 22 hpf in the gut endoderm and at 40 hpf expanded to the newly formed liver bud. Endodermal nav3a expression was controlled by Wnt2bb signaling and was independent of FGF and BMP signaling. Morpholino-mediated knockdown of nav3a resulted in a significantly reduced liver size, and impaired development of pancreas and swim bladder. In vivo time-lapse imaging of liver development in nav3a morphants revealed a failure of hepatoblast movement out from the gut endoderm during the liver budding stage, with hepatoblasts being retained in the intestinal endoderm. In hepatocytes in vitro, nav3a acts as a positive modulator of actin assembly in lamellipodia and filipodia extensions, allowing cellular movement. Knockdown of nav3a in vitro impeded hepatocyte movement. Endodermal-specific overexpression of nav3a in vivo resulted in additional ectopic endodermal budding beyond the normal liver and pancreatic budding sites. We conclude that nav3a is required for directing endodermal organogenesis involving coordination of endodermal cell behavior.


Subject(s)
Liver/embryology , Liver/metabolism , Nerve Tissue Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Base Sequence , DNA Primers/genetics , Endoderm/embryology , Endoderm/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , In Situ Hybridization , Intestinal Mucosa/metabolism , Intestines/embryology , Liver/abnormalities , Liver/innervation , Models, Biological , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Organogenesis/genetics , Organogenesis/physiology , RNA, Antisense/genetics , Signal Transduction , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/genetics , Wnt Proteins/metabolism , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
18.
Circ Res ; 110(4): 578-87, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22247485

ABSTRACT

RATIONALE: The importance for Bmp signaling during embryonic stem cell differentiation into myocardial cells has been recognized. The question when and where Bmp signaling in vivo regulates myocardial differentiation has remained largely unanswered. OBJECTIVE: To identify when and where Bmp signaling regulates cardiogenic differentiation. METHODS AND RESULTS: Here we have observed that in zebrafish embryos, Bmp signaling is active in cardiac progenitor cells prior to their differentiation into cardiomyocytes. Bmp signaling is continuously required during somitogenesis within the anterior lateral plate mesoderm to induce myocardial differentiation. Surprisingly, Bmp signaling is actively repressed in differentiating myocardial cells. We identified the inhibitory Smad6a, which is expressed in the cardiac tissue, to be required to inhibit Bmp signaling and thereby promote expansion of the ventricular myocardium. CONCLUSION: Bmp signaling exerts opposing effects on myocardial differentiation in the embryo by promoting as well as inhibiting cardiac growth.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Embryonic Stem Cells/metabolism , Heart/embryology , Myocytes, Cardiac/metabolism , Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Animals , Animals, Genetically Modified , Body Patterning , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/genetics , Cell Proliferation , Gene Expression Regulation, Developmental , Genes, Reporter , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Mutation , Smad6 Protein/metabolism , T-Box Domain Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
19.
J Clin Invest ; 134(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38747293

ABSTRACT

Molecular characterization of vascular anomalies has revealed that affected endothelial cells (ECs) harbor gain-of-function (GOF) mutations in the gene encoding the catalytic α subunit of PI3Kα (PIK3CA). These PIK3CA mutations are known to cause solid cancers when occurring in other tissues. PIK3CA-related vascular anomalies, or "PIKopathies," range from simple, i.e., restricted to a particular form of malformation, to complex, i.e., presenting with a range of hyperplasia phenotypes, including the PIK3CA-related overgrowth spectrum. Interestingly, development of PIKopathies is affected by fluid shear stress (FSS), a physiological stimulus caused by blood or lymph flow. These findings implicate PI3K in mediating physiological EC responses to FSS conditions characteristic of lymphatic and capillary vessel beds. Consistent with this hypothesis, increased PI3K signaling also contributes to cerebral cavernous malformations, a vascular disorder that affects low-perfused brain venous capillaries. Because the GOF activity of PI3K and its signaling partners are excellent drug targets, understanding PIK3CA's role in the development of vascular anomalies may inform therapeutic strategies to normalize EC responses in the diseased state. This Review focuses on PIK3CA's role in mediating EC responses to FSS and discusses current understanding of PIK3CA dysregulation in a range of vascular anomalies that particularly affect low-perfused regions of the vasculature. We also discuss recent surprising findings linking increased PI3K signaling to fast-flow arteriovenous malformations in hereditary hemorrhagic telangiectasias.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Vascular Malformations , Humans , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Animals , Vascular Malformations/genetics , Vascular Malformations/pathology , Vascular Malformations/physiopathology , Vascular Malformations/metabolism , Vascular Malformations/enzymology , Endothelial Cells/enzymology , Endothelial Cells/pathology , Endothelial Cells/metabolism , Stress, Mechanical , Gain of Function Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/metabolism , Hemangioma, Cavernous, Central Nervous System/physiopathology , Hemangioma, Cavernous, Central Nervous System/pathology
20.
J Invest Dermatol ; 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38431221

ABSTRACT

Common capillary malformations are red vascular skin lesions, most commonly associated with somatic activating GNAQ or GNA11 mutations. We focused on capillary malformations lacking such a mutation to identify previously unreported genetic causes. We used targeted next-generation sequencing on 82 lesions. Bioinformatic analysis allowed the identification of 9 somatic pathogenic variants in PIK3R1 and PIK3CA, encoding for the regulatory and catalytic subunits of phosphoinositide 3-kinase, respectively. Recharacterization of these lesions unraveled a common phenotype: a pale capillary malformation associated with visible dilated veins. Primary endothelial cells from 2 PIK3R1-mutated lesions were isolated, and PI3k-Akt-mTOR and RAS-RAF-MAPK signaling were assessed by western blot. This unveiled an abnormal increase in Akt phosphorylation, effectively reduced by PI3K pathway inhibitors, such as mTOR, Akt, and PIK3CA inhibitors. The effects of mutant PIK3R1 were further studied using zebrafish embryos. Endothelium-specific expression of PIK3R1 mutants resulted in abnormal development of the posterior capillary-venous plexus. In summary, capillary malformation associated with visible dilated veins emerges as a clinical entity associated with somatic pathogenic variants in PIK3R1 or PIK3CA (nonhotspot). Our findings suggest that the activated Akt signaling can be effectively reversed by PI3K pathway inhibitors. In addition, the proposed zebrafish model holds promise as a valuable tool for future drug screening aimed at developing patient-tailored treatments.

SELECTION OF CITATIONS
SEARCH DETAIL