Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Development ; 148(9)2021 05 01.
Article in English | MEDLINE | ID: mdl-33969874

ABSTRACT

During heart development, epicardial cells residing within the outer layer undergo epithelial-mesenchymal transition (EMT) and migrate into the underlying myocardium to support organ growth and morphogenesis. Disruption of epicardial EMT results in embryonic lethality, yet its regulation is poorly understood. Here, we report epicardial EMT within the mesothelial layer of the mouse embryonic heart at ultra-high resolution using scanning electron microscopy combined with immunofluorescence analyses. We identified morphologically active EMT regions that associated with key components of the extracellular matrix, including the basement membrane-associated proteoglycan agrin. Deletion of agrin resulted in impaired EMT and compromised development of the epicardium, accompanied by downregulation of Wilms' tumor 1. Agrin enhanced EMT in human embryonic stem cell-derived epicardial-like cells by decreasing ß-catenin and promoting pFAK localization at focal adhesions, and promoted the aggregation of dystroglycan within the Golgi apparatus in murine epicardial cells. Loss of agrin resulted in dispersal of dystroglycan in vivo, disrupting basement membrane integrity and impairing EMT. Our results provide new insights into the role of the extracellular matrix in heart development and implicate agrin as a crucial regulator of epicardial EMT.


Subject(s)
Agrin/metabolism , Epithelial-Mesenchymal Transition/physiology , Extracellular Matrix Proteins/metabolism , Heart/embryology , Heart/growth & development , Organogenesis/physiology , Animals , Female , Genetic Heterogeneity , Golgi Apparatus , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Pericardium/metabolism , beta Catenin/genetics , beta Catenin/metabolism
2.
Nature ; 557(7705): 439-445, 2018 05.
Article in English | MEDLINE | ID: mdl-29743679

ABSTRACT

In vertebrate hearts, the ventricular trabecular myocardium develops as a sponge-like network of cardiomyocytes that is critical for contraction and conduction, ventricular septation, papillary muscle formation and wall thickening through the process of compaction 1 . Defective trabeculation leads to embryonic lethality2-4 or non-compaction cardiomyopathy (NCC) 5 . There are divergent views on when and how trabeculation is initiated in different species. In zebrafish, trabecular cardiomyocytes extrude from compact myocardium 6 , whereas in chicks, chamber wall thickening occurs before overt trabeculation 7 . In mice, the onset of trabeculation has not been described, but is proposed to begin at embryonic day 9.0, when cardiomyocytes form radially oriented ribs 2 . Endocardium-myocardium communication is essential for trabeculation, and numerous signalling pathways have been identified, including Notch2,8 and Neuregulin (NRG) 4 . Late disruption of the Notch pathway causes NCC 5 . Whereas it has been shown that mutations in the extracellular matrix (ECM) genes Has2 and Vcan prevent the formation of trabeculae in mice9,10 and the matrix metalloprotease ADAMTS1 promotes trabecular termination 3 , the pathways involved in ECM dynamics and the molecular regulation of trabeculation during its early phases remain unexplored. Here we present a model of trabeculation in mice that integrates dynamic endocardial and myocardial cell behaviours and ECM remodelling, and reveal new epistatic relationships between the involved signalling pathways. NOTCH1 signalling promotes ECM degradation during the formation of endocardial projections that are critical for individualization of trabecular units, whereas NRG1 promotes myocardial ECM synthesis, which is necessary for trabecular rearrangement and growth. These systems interconnect through NRG1 control of Vegfa, but act antagonistically to establish trabecular architecture. These insights enabled the prediction of persistent ECM and cardiomyocyte growth in a mouse NCC model, providing new insights into the pathophysiology of congenital heart disease.


Subject(s)
Heart/embryology , Myocardium/cytology , Myocardium/metabolism , Neuregulin-1/metabolism , Organogenesis , Receptor, Notch1/metabolism , Animals , Disease Models, Animal , Endocardium/cytology , Endocardium/metabolism , Extracellular Matrix/metabolism , Heart Diseases/congenital , Heart Diseases/metabolism , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Neuregulin-1/genetics , Receptor, Notch1/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
3.
Nature ; 547(7662): 179-184, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28581497

ABSTRACT

The adult mammalian heart is non-regenerative owing to the post-mitotic nature of cardiomyocytes. The neonatal mouse heart can regenerate, but only during the first week of life. Here we show that changes in the composition of the extracellular matrix during this week can affect cardiomyocyte growth and differentiation in mice. We identify agrin, a component of neonatal extracellular matrix, as required for the full regenerative capacity of neonatal mouse hearts. In vitro, recombinant agrin promotes the division of cardiomyocytes that are derived from mouse and human induced pluripotent stem cells through a mechanism that involves the disassembly of the dystrophin-glycoprotein complex, and Yap- and ERK-mediated signalling. In vivo, a single administration of agrin promotes cardiac regeneration in adult mice after myocardial infarction, although the degree of cardiomyocyte proliferation observed in this model suggests that there are additional therapeutic mechanisms. Together, our results uncover a new inducer of mammalian heart regeneration and highlight fundamental roles of the extracellular matrix in cardiac repair.


Subject(s)
Agrin/metabolism , Extracellular Matrix Proteins/metabolism , Heart/physiology , Regeneration , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Cell Cycle Proteins , Cell Proliferation , Dystroglycans/metabolism , Female , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , YAP-Signaling Proteins
4.
Circulation ; 142(9): 868-881, 2020 09.
Article in English | MEDLINE | ID: mdl-32508131

ABSTRACT

BACKGROUND: Ischemic heart diseases are leading causes of death and reduced life quality worldwide. Although revascularization strategies significantly reduce mortality after acute myocardial infarction (MI), a large number of patients with MI develop chronic heart failure over time. We previously reported that a fragment of the extracellular matrix protein agrin promotes cardiac regeneration after MI in adult mice. METHODS: To test the therapeutic potential of agrin in a preclinical porcine model, we performed ischemia-reperfusion injuries using balloon occlusion for 60 minutes followed by a 3-, 7-, or 28-day reperfusion period. RESULTS: We demonstrated that local (antegrade) delivery of recombinant human agrin to the infarcted pig heart can target the affected regions in an efficient and clinically relevant manner. A single dose of recombinant human agrin improved heart function, infarct size, fibrosis, and adverse remodeling parameters 28 days after MI. Short-term MI experiments along with complementary murine studies revealed myocardial protection, improved angiogenesis, inflammatory suppression, and cell cycle reentry as agrin's mechanisms of action. CONCLUSIONS: A single dose of agrin is capable of reducing ischemia-reperfusion injury and improving heart function, demonstrating that agrin could serve as a therapy for patients with acute MI and potentially heart failure.


Subject(s)
Agrin/pharmacology , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Recovery of Function/drug effects , Animals , Humans , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Recombinant Proteins/pharmacology , Swine
5.
Nature ; 520(7548): 466-73, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25903628

ABSTRACT

It has been more than 30 years since the publication of the new head hypothesis, which proposed that the vertebrate head is an evolutionary novelty resulting from the emergence of neural crest and cranial placodes. Neural crest generates the skull and associated connective tissues, whereas placodes produce sensory organs. However, neither crest nor placodes produce head muscles, which are a crucial component of the complex vertebrate head. We discuss emerging evidence for a surprising link between the evolution of head muscles and chambered hearts - both systems arise from a common pool of mesoderm progenitor cells within the cardiopharyngeal field of vertebrate embryos. We consider the origin of this field in non-vertebrate chordates and its evolution in vertebrates.


Subject(s)
Biological Evolution , Branchial Region/embryology , Head/anatomy & histology , Head/embryology , Heart/anatomy & histology , Heart/embryology , Vertebrates/anatomy & histology , Vertebrates/embryology , Animals , Branchial Region/anatomy & histology , Branchial Region/cytology , Mesoderm/cytology , Models, Biological , Muscles/anatomy & histology , Muscles/cytology , Muscles/embryology , Neural Crest/cytology
7.
Carcinogenesis ; 38(4): 359-366, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28334384

ABSTRACT

Regeneration in mammals is restricted to distinct tissues and occurs mainly by expansion and maturation of resident stem cells. During regeneration, even subtle mutations in the proliferating cells may cause a detrimental effect by eliciting abnormal differentiation or malignant transformation. Indeed, cancer in mammals has been shown to arise through deregulation of stem cells maturation, which often leads to a differentiation block and cell transformation. In contrast, lower organisms such as amphibians retain a remarkable regenerative capacity in various organs, which occurs via de- and re-differentiation of mature cells. Interestingly, regenerating amphibian cells are highly resistant to oncogenic transformation. Therapeutic approaches to improve mammalian regeneration mainly include stem-cell transplantations; but, these have proved unsuccessful in non-regenerating organs such as the heart. A recently developed approach is to induce de-differentiation of mature cardiomyocytes using factors that trigger their re-entry into the cell cycle. This novel approach raises numerous questions regarding the balance between transformation and regeneration induced by de-differentiation of mature mammalian somatic cells. Can this balance be controlled artificially? Do de-differentiated cells acquire the protection mechanisms seen in regenerating cells of lower organisms? Is this model unique to the cardiac tissue, which rarely develops tumors? This review describes regeneration processes in both mammals and lower organisms and, particularly, the ability of regenerating cells to avoid transformation. By comparing the characteristics of mammalian embryonic and somatic cells, we discuss therapeutic strategies of using various cell populations for regeneration. Finally, we describe a novel cardiac regeneration approach and its implications for regenerative medicine.


Subject(s)
Amphibians/physiology , Mammals/physiology , Neoplasms/pathology , Regeneration/physiology , Animals , Cell Differentiation/physiology , Cell Transformation, Neoplastic/pathology , Heart/physiology , Humans , Stem Cells/physiology
8.
Development ; 141(13): 2611-20, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24924195

ABSTRACT

The transition between the proliferation and differentiation of progenitor cells is a key step in organogenesis, and alterations in this process can lead to developmental disorders. The extracellular signal-regulated kinase 1/2 (ERK) signaling pathway is one of the most intensively studied signaling mechanisms that regulates both proliferation and differentiation. How a single molecule (e.g. ERK) can regulate two opposing cellular outcomes is still a mystery. Using both chick and mouse models, we shed light on the mechanism responsible for the switch from proliferation to differentiation of head muscle progenitors and implicate ERK subcellular localization. Manipulation of the fibroblast growth factor (FGF)-ERK signaling pathway in chick embryos in vitro and in vivo demonstrated that blockage of this pathway accelerated myogenic differentiation, whereas its activation diminished it. We next examined whether the spatial subcellular localization of ERK could act as a switch between proliferation (nuclear ERK) and differentiation (cytoplasmic ERK) of muscle progenitors. A myristoylated peptide that blocks importin 7-mediated ERK nuclear translocation induced robust myogenic differentiation of muscle progenitor/stem cells in both head and trunk. In the mouse, analysis of Sprouty mutant embryos revealed that increased ERK signaling suppressed both head and trunk myogenesis. Our findings, corroborated by mathematical modeling, suggest that ERK shuttling between the nucleus and the cytoplasm provides a switch-like transition between proliferation and differentiation of muscle progenitors.


Subject(s)
Cell Differentiation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Muscle Development/physiology , Stem Cells/physiology , Active Transport, Cell Nucleus/physiology , Animals , Bromodeoxyuridine , Cell Proliferation , Chick Embryo , DNA Primers/genetics , Fluorescent Antibody Technique , Mice , Mice, Transgenic , Models, Biological , Real-Time Polymerase Chain Reaction
10.
Proc Natl Acad Sci U S A ; 109(28): 11211-6, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22736793

ABSTRACT

A fundamental aspect of skeletal myogenesis involves extensive rounds of cell fusion, in which individual myoblasts are incorporated into growing muscle fibers. Here we demonstrate that N-WASp, a ubiquitous nucleation-promoting factor of branched microfilament arrays, is an essential contributor to skeletal muscle-cell fusion in developing mouse embryos. Analysis both in vivo and in primary satellite-cell cultures, shows that disruption of N-WASp function does not interfere with the program of skeletal myogenic differentiation, and does not affect myoblast motility, morphogenesis and attachment capacity. N-WASp-deficient myoblasts, however, fail to fuse. Furthermore, our analysis suggests that myoblast fusion requires N-WASp activity in both partners of a fusing myoblast pair. These findings reveal a specific role for N-WASp during mammalian myogenesis. WASp-family elements appear therefore to act as universal mediators of the myogenic cell-cell fusion mechanism underlying formation of functional muscle fibers, in both vertebrate and invertebrate species.


Subject(s)
Actins/metabolism , Gene Expression Regulation, Developmental , Muscles/cytology , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Animals , Cell Differentiation , Cell Fusion , Cells, Cultured , Crosses, Genetic , Drosophila , Heterozygote , Mice , Mice, Inbred ICR , Models, Biological , Muscle Development , Muscles/embryology , Time Factors
11.
Proc Natl Acad Sci U S A ; 109(46): 18839-44, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23112163

ABSTRACT

The search for developmental mechanisms driving vertebrate organogenesis has paved the way toward a deeper understanding of birth defects. During embryogenesis, parts of the heart and craniofacial muscles arise from pharyngeal mesoderm (PM) progenitors. Here, we reveal a hierarchical regulatory network of a set of transcription factors expressed in the PM that initiates heart and craniofacial organogenesis. Genetic perturbation of this network in mice resulted in heart and craniofacial muscle defects, revealing robust cross-regulation between its members. We identified Lhx2 as a previously undescribed player during cardiac and pharyngeal muscle development. Lhx2 and Tcf21 genetically interact with Tbx1, the major determinant in the etiology of DiGeorge/velo-cardio-facial/22q11.2 deletion syndrome. Furthermore, knockout of these genes in the mouse recapitulates specific cardiac features of this syndrome. We suggest that PM-derived cardiogenesis and myogenesis are network properties rather than properties specific to individual PM members. These findings shed new light on the developmental underpinnings of congenital defects.


Subject(s)
Body Patterning/physiology , Embryo, Mammalian/embryology , Head/embryology , Heart/embryology , Mesoderm/embryology , Muscle, Skeletal/embryology , Myocardium , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout
12.
Development ; 138(21): 4777-87, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21989917

ABSTRACT

The embryonic heart is composed of two cell layers: the myocardium, which contributes to cardiac muscle tissue, and the endocardium, which covers the inner lumen of the heart. Whereas significant progress has been made toward elucidating the embryonic origins of the myocardium, the origins of the endocardium remain unclear. Here, we have identified an endocardium-forming field medial to the cardiac crescent, in a continuum with the endothelial plexus. In vivo live imaging of quail embryos revealed that endothelial progenitors, like second/anterior heart field progenitors, migrate to, and enter, the heart from the arterial pole. Furthermore, embryonic endothelial cells implanted into the cardiac crescent contribute to the endocardium, but not to the myocardium. In mouse, lineage analysis focusing on endocardial cells revealed an unexpected heterogeneity in the origins of the endocardium. To gain deeper insight into this heterogeneity, we conditionally ablated Flk1 in distinct cardiovascular progenitor populations; FLK1 is required in vivo for formation of the endocardium in the Mesp1 and Tie2 lineages, but not in the Isl1 lineage. Ablation of Flk1 coupled with lineage analysis in the Isl1 lineage revealed that endothelium-derived Isl1(-) endocardial cells were significantly increased, whereas Isl1(+) endocardial cells were reduced, suggesting that the endocardium is capable of undergoing regulative compensatory growth. Collectively, our findings demonstrate that the second heart field contains distinct myocardial and endocardial progenitor populations. We suggest that the endocardium derives, at least in part, from vascular endothelial cells.


Subject(s)
Endocardium/embryology , Endothelial Cells/physiology , Heart/anatomy & histology , Heart/embryology , Stem Cells/physiology , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Lineage/physiology , Chick Embryo , Chimera , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/physiology , Endocardium/cytology , Endothelial Cells/cytology , Gene Knockdown Techniques , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Myocardium/cytology , Quail , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
Development ; 138(9): 1827-38, 2011 May.
Article in English | MEDLINE | ID: mdl-21447558

ABSTRACT

Neural crest development involves epithelial-mesenchymal transition (EMT), during which epithelial cells are converted into individual migratory cells. Notably, the same signaling pathways regulate EMT function during both development and tumor metastasis. p53 plays multiple roles in the prevention of tumor development; however, its precise roles during embryogenesis are less clear. We have investigated the role of p53 in early cranial neural crest (CNC) development in chick and mouse embryos. In the mouse, p53 knockout embryos displayed broad craniofacial defects in skeletal, neuronal and muscle tissues. In the chick, p53 is expressed in CNC progenitors and its expression decreases with their delamination from the neural tube. Stabilization of p53 protein using a pharmacological inhibitor of its negative regulator, MDM2, resulted in reduced SNAIL2 (SLUG) and ETS1 expression, fewer migrating CNC cells and in craniofacial defects. By contrast, electroporation of a dominant-negative p53 construct increased PAX7(+) SOX9(+) CNC progenitors and EMT/delamination of CNC from the neural tube, although the migration of these cells to the periphery was impaired. Investigating the underlying molecular mechanisms revealed that p53 coordinates CNC cell growth and EMT/delamination processes by affecting cell cycle gene expression and proliferation at discrete developmental stages; disruption of these processes can lead to craniofacial defects.


Subject(s)
Cell Proliferation , Epithelial-Mesenchymal Transition/genetics , Laminin/metabolism , Neural Crest/embryology , Skull/embryology , Tumor Suppressor Protein p53/physiology , Animals , Cells, Cultured , Chick Embryo , Craniofacial Abnormalities/complications , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Embryo, Mammalian , Epithelial-Mesenchymal Transition/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Musculoskeletal Abnormalities/complications , Musculoskeletal Abnormalities/embryology , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/pathology , Neural Crest/cytology , Neural Crest/metabolism , Skull/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
14.
Cell Rep ; 43(5): 114162, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38678558

ABSTRACT

Zebrafish have a lifelong cardiac regenerative ability after damage, whereas mammals lose this capacity during early postnatal development. This study investigated whether the declining expression of growth factors during postnatal mammalian development contributes to the decrease of cardiomyocyte regenerative potential. Besides confirming the proliferative ability of neuregulin 1 (NRG1), interleukin (IL)1b, receptor activator of nuclear factor kappa-Β ligand (RANKL), insulin growth factor (IGF)2, and IL6, we identified other potential pro-regenerative factors, with BMP7 exhibiting the most pronounced efficacy. Bmp7 knockdown in neonatal mouse cardiomyocytes and loss-of-function in adult zebrafish during cardiac regeneration reduced cardiomyocyte proliferation, indicating that Bmp7 is crucial in the regenerative stages of mouse and zebrafish hearts. Conversely, bmp7 overexpression in regenerating zebrafish or administration at post-mitotic juvenile and adult mouse stages, in vitro and in vivo following myocardial infarction, enhanced cardiomyocyte cycling. Mechanistically, BMP7 stimulated proliferation through BMPR1A/ACVR1 and ACVR2A/BMPR2 receptors and downstream SMAD5, ERK, and AKT signaling. Overall, BMP7 administration is a promising strategy for heart regeneration.


Subject(s)
Bone Morphogenetic Protein 7 , Cell Proliferation , Myocytes, Cardiac , Regeneration , Zebrafish , Animals , Female , Male , Mice , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/genetics , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Neuregulin-1/metabolism , Neuregulin-1/genetics , Signal Transduction , Smad5 Protein/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
15.
Development ; 137(18): 2989-3000, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20702560

ABSTRACT

The anterior heart field (AHF) encompasses a niche in which mesoderm-derived cardiac progenitors maintain their multipotent and undifferentiated nature in response to signals from surrounding tissues. Here, we investigate the signaling mechanism that promotes the shift from proliferating cardiac progenitors to differentiating cardiomyocytes in chick embryos. Genomic and systems biology approaches, as well as perturbations of signaling molecules, in vitro and in vivo, reveal tight crosstalk between the bone morphogenetic protein (BMP) and fibroblast growth factor (FGF) signaling pathways within the AHF niche: BMP4 promotes myofibrillar gene expression and cardiomyocyte contraction by blocking FGF signaling. Furthermore, inhibition of the FGF-ERK pathway is both sufficient and necessary for these processes, suggesting that FGF signaling blocks premature differentiation of cardiac progenitors in the AHF. We further revealed that BMP4 induced a set of neural crest-related genes, including MSX1. Overexpression of Msx1 was sufficient to repress FGF gene expression and cell proliferation, thereby promoting cardiomyocyte differentiation. Finally, we show that BMP-induced cardiomyocyte differentiation is diminished following cranial neural crest ablation, underscoring the key roles of these cells in the regulation of AHF cell differentiation. Hence, BMP and FGF signaling pathways act via inter- and intra-regulatory loops in multiple tissues, to coordinate the balance between proliferation and differentiation of cardiac progenitors.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Cell Differentiation , Heart/embryology , Myocytes, Cardiac/metabolism , Signal Transduction , Stem Cells/metabolism , Animals , Bone Morphogenetic Protein 4/genetics , Cell Proliferation , Chick Embryo , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Heart/physiology , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Myocytes, Cardiac/cytology , Stem Cells/cytology , Tissue Culture Techniques
16.
Development ; 137(17): 2961-71, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20699298

ABSTRACT

In vertebrates, body musculature originates from somites, whereas head muscles originate from the cranial mesoderm. Neck muscles are located in the transition between these regions. We show that the chick occipital lateral plate mesoderm has myogenic capacity and gives rise to large muscles located in the neck and thorax. We present molecular and genetic evidence to show that these muscles not only have a unique origin, but additionally display a distinct temporal development, forming later than any other muscle group described to date. We further report that these muscles, found in the body of the animal, develop like head musculature rather than deploying the programme used by the trunk muscles. Using mouse genetics we reveal that these muscles are formed in trunk muscle mutants but are absent in head muscle mutants. In concordance with this conclusion, their connective tissue is neural crest in origin. Finally, we provide evidence that the mechanism by which these neck muscles develop is conserved in vertebrates.


Subject(s)
Mesoderm/embryology , Muscle Development , Neck Muscles/embryology , Animals , Animals, Genetically Modified , Avian Proteins/genetics , Biological Evolution , Chick Embryo , Coturnix , Gene Expression Regulation, Developmental , Mice , Muscle Development/genetics , Mutation , Neural Crest/embryology , Paired Box Transcription Factors/genetics , Somites/embryology , Transplantation Chimera/embryology , Transplantation Chimera/genetics
17.
Nat Commun ; 14(1): 5810, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37726308

ABSTRACT

The tumor microenvironment (TME) is comprised of non-malignant cells that interact with each other and with cancer cells, critically impacting cancer biology. The TME is complex, and understanding it requires simplifying approaches. Here we provide an experimental-mathematical approach to decompose the TME into small circuits of interacting cell types. We find, using female breast cancer single-cell-RNA-sequencing data, a hierarchical network of interactions, with cancer-associated fibroblasts (CAFs) at the top secreting factors primarily to tumor-associated macrophages (TAMs). This network is composed of repeating circuit motifs. We isolate the strongest two-cell circuit motif by culturing fibroblasts and macrophages in-vitro, and analyze their dynamics and transcriptomes. This isolated circuit recapitulates the hierarchy of in-vivo interactions, and enables testing the effect of ligand-receptor interactions on cell dynamics and function, as we demonstrate by identifying a mediator of CAF-TAM interactions - RARRES2, and its receptor CMKLR1. Thus, the complexity of the TME may be simplified by identifying small circuits, facilitating the development of strategies to modulate the TME.


Subject(s)
Cancer-Associated Fibroblasts , Tumor Microenvironment , Female , Humans , Fibroblasts , Biological Transport , Cell Communication
18.
Nat Cardiovasc Res ; 2(4): 383-398, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37974970

ABSTRACT

Cardiomyocyte proliferation and dedifferentiation have fueled the field of regenerative cardiology in recent years, whereas the reverse process of redifferentiation remains largely unexplored. Redifferentiation is characterized by the restoration of function lost during dedifferentiation. Previously, we showed that ERBB2-mediated heart regeneration has these two distinct phases: transient dedifferentiation and redifferentiation. Here we survey the temporal transcriptomic and proteomic landscape of dedifferentiation-redifferentiation in adult mouse hearts and reveal that well-characterized dedifferentiation features largely return to normal, although elements of residual dedifferentiation remain, even after the contractile function is restored. These hearts appear rejuvenated and show robust resistance to ischemic injury, even 5 months after redifferentiation initiation. Cardiomyocyte redifferentiation is driven by negative feedback signaling and requires LATS1/2 Hippo pathway activity. Our data reveal the importance of cardiomyocyte redifferentiation in functional restoration during regeneration but also protection against future insult, in what could lead to a potential prophylactic treatment against ischemic heart disease for at-risk patients.

19.
Science ; 380(6646): 758-764, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37200435

ABSTRACT

Zebrafish hearts can regenerate by replacing damaged tissue with new cardiomyocytes. Although the steps leading up to the proliferation of surviving cardiomyocytes have been extensively studied, little is known about the mechanisms that control proliferation and redifferentiation to a mature state. We found that the cardiac dyad, a structure that regulates calcium handling and excitation-contraction coupling, played a key role in the redifferentiation process. A component of the cardiac dyad called leucine-rich repeat-containing 10 (Lrrc10) acted as a negative regulator of proliferation, prevented cardiomegaly, and induced redifferentiation. We found that its function was conserved in mammalian cardiomyocytes. This study highlights the importance of the underlying mechanisms required for heart regeneration and their application to the generation of fully functional cardiomyocytes.


Subject(s)
Calcium , Heart , Myocytes, Cardiac , Regeneration , Sarcomeres , Zebrafish , Animals , Calcium/physiology , Cell Proliferation , Heart/physiology , Myocytes, Cardiac/physiology , Sarcomeres/physiology , Zebrafish/physiology
20.
Cell Stem Cell ; 30(1): 96-111.e6, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36516837

ABSTRACT

The efficacy and safety of gene-therapy strategies for indications like tissue damage hinge on precision; yet, current methods afford little spatial or temporal control of payload delivery. Here, we find that tissue-regeneration enhancer elements (TREEs) isolated from zebrafish can direct targeted, injury-associated gene expression from viral DNA vectors delivered systemically in small and large adult mammalian species. When employed in combination with CRISPR-based epigenome editing tools in mice, zebrafish TREEs stimulated or repressed the expression of endogenous genes after ischemic myocardial infarction. Intravenously delivered recombinant AAV vectors designed with a TREE to direct a constitutively active YAP factor boosted indicators of cardiac regeneration in mice and improved the function of the injured heart. Our findings establish the application of contextual enhancer elements as a potential therapeutic platform for spatiotemporally controlled tissue regeneration in mammals.


Subject(s)
Enhancer Elements, Genetic , Genetic Therapy , Heart , Myocardial Infarction , Myocytes, Cardiac , Regeneration , Animals , Mice , Cell Proliferation , Heart/physiology , Myocardial Infarction/genetics , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Zebrafish/genetics , Genetic Therapy/methods , Regeneration/genetics
SELECTION OF CITATIONS
SEARCH DETAIL